1
|
Ejlalidiz M, Mehri-Ghahfarrokhi A, Saberiyan M. Identification of hub genes and pathways in Uterine corpus endometrial carcinoma (UCEC): A comprehensive in silico study. Biochem Biophys Rep 2024; 40:101860. [PMID: 39552710 PMCID: PMC11565547 DOI: 10.1016/j.bbrep.2024.101860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 10/27/2024] [Indexed: 11/19/2024] Open
Abstract
Background Uterine corpus endometrial carcinoma (UCEC), derived from the endometrium, is the most common type of endometrial malignasis. This gynecological malignancy is very common all over the world, especially in developed countries and shows a potentially rising trend correlated with the increase in obese women. Methods Differentially Expressed Genes (DEGs) analysis was conducted on GSE7305 and GSE25628 datasets from the Gene Expression Omnibus (GEO). DEGs were identified using GEO2R (adjusted p-value <0.05, |logFC| > 1). Pathway analysis employed KEGG and Gene Ontology databases, while protein-protein interactions were analyzed using Cytoscape and Gephi. GEPIA was used for target gene validation. Results We have identified 304 common DEGs and 78 hub genes using GEO and PPI analysis, respectively. The GO and KEGG pathways analysis revealed enrichment of DEGs in extracellular matrix structural constituent, extracellular space, cell adhesion, and ECM-receptor interaction. GEPIA analysis identified three genes, ENG, GNG4, and ECT2, whose expression significantly differed between normal and tumor samples. Conclusion This analysis study identified the hub genes and associated pathways involved in the pathogenesis of UCEC. The identified hub genes exhibit remarkable potential as diagnostic biomarkers, providing a significant opportunity for early diagnosis and more effective therapeutic approaches for UCEC.
Collapse
Affiliation(s)
- Mahsa Ejlalidiz
- Medical Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ameneh Mehri-Ghahfarrokhi
- Clinical Research Developmental Unit, Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammadreza Saberiyan
- Department of Medical Genetics, School of Medical Sciences, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
2
|
Yao Y, Yang K, Wang Q, Zhu Z, Li S, Li B, Feng B, Tang C. Prediction of CAF-related genes in immunotherapy and drug sensitivity in hepatocellular carcinoma: a multi-database analysis. Genes Immun 2024; 25:55-65. [PMID: 38233508 PMCID: PMC10873201 DOI: 10.1038/s41435-024-00252-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 01/19/2024]
Abstract
This study aims to identify the cancer-associated fibroblasts (CAF)-related genes that can affect immunotherapy and drug sensitivity in hepatocellular carcinoma (HCC). Expression data and survival data associated with HCC were obtained in The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Weighted correlation network analysis (WGCNA) analysis was performed to obtain CAF-related genes. Least Absolute Shrinkage and Selection Operator (LASSO) regression was used for regression analysis and risk models. Subsequently, Gene Set Enrichment Analysis (GSEA) analysis, Gene Set Enrichment Analysis (ssGSEA) analysis, Tumor Immune Dysfunction and Exclusion (TIDE) analysis and drug sensitivity analysis were performed on the risk models. Survival analysis of CAF scores showed that the survival rate was lower in samples with high CAF scores than those with low scores. However, this difference was not significant, suggesting CAF may not directly influence the prognosis of HCC patients. Further screening of CAF-related genes yielded 33 CAF-related genes. Seven risk models constructed based on CDR2L, SPRED1, PFKP, ENG, KLF2, FSCN1 and VCAN, showed significant differences in immunotherapy and partial drug sensitivity in HCC. Seven CAF-related genes may have important roles in immunotherapy, drug sensitivity and prognostic survival in HCC patients.
Collapse
Affiliation(s)
- Yi Yao
- Division 1, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - KaiQing Yang
- Division 1, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Qiang Wang
- Division 1, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Zeming Zhu
- Division 2, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Sheng Li
- Division 1, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Bin Li
- Division 1, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Bin Feng
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China.
| | - Caixi Tang
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China.
| |
Collapse
|
3
|
Oladejo M, Nguyen HM, Wood L. CD105 in the progression and therapy of renal cell carcinoma. Cancer Lett 2023; 570:216327. [PMID: 37499740 DOI: 10.1016/j.canlet.2023.216327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023]
Abstract
Molecular biomarkers that interact with the vascular and immune compartments play an important role in the progression of solid malignancies. CD105, which is a component of the transforming growth factor beta (TGF β) signaling cascade, has long been studied for its role in potentiating angiogenesis in numerous cancers. In renal cell carcinoma (RCC), the role of CD105 is more complicated due to its diverse expression profile on the tumor cells, tumor vasculature, and the components of the immune system. Since its discovery, its angiogenic role has overshadowed other potential functions, especially in cancers. In this review, we aim to summarize the recent evidence and findings of the multifunctional roles of CD105 in angiogenesis and immunomodulation in the context of the various subtypes of RCC, with a specific emphasis on the clear cell RCC subtype. Since CD105 is an established biomarker and tumor antigen, we also provide an update on the preclinical and clinical applications of CD105 as a therapeutic platform in RCC.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Hong-My Nguyen
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Laurence Wood
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA.
| |
Collapse
|
4
|
Afonso J, Gonçalves C, Costa M, Ferreira D, Santos L, Longatto-Filho A, Baltazar F. Glucose Metabolism Reprogramming in Bladder Cancer: Hexokinase 2 (HK2) as Prognostic Biomarker and Target for Bladder Cancer Therapy. Cancers (Basel) 2023; 15:cancers15030982. [PMID: 36765947 PMCID: PMC9913750 DOI: 10.3390/cancers15030982] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Proliferating cancer cells are able to reprogram their energy metabolism, favouring glycolysis even in the presence of oxygen and fully functioning mitochondria. Research is needed to validate the glycolysis-related proteins as prognostic/predictive biomarkers in urothelial bladder carcinoma (UBC), a malignancy tagged by high recurrence rates and poor response to chemotherapy. Here, we assessed GLUT1, HK2, PFKL, PKM2, phospho-PDH, and LDHA immunoexpression in 76 UBC samples, differentiating among urothelial, fibroblast, and endothelial cells and among normoxic versus hypoxic areas. We additionally studied the functional effects of the HK2 inhibitor 2-deoxy-D-glucose (2DG) in "in vitro" and "in vivo" preclinical UBC models. We showed that the expression of the glycolysis-related proteins is associated with UBC aggressiveness and poor prognosis. HK2 remained as an independent prognostic factor for disease-free and overall survival. 2DG decreased the UBC cell's viability, proliferation, migration, and invasion; the inhibition of cell cycle progression and apoptosis occurrence was also verified. A significant reduction in tumour growth and blood vessel formation upon 2DG treatment was observed in the chick chorioallantoic membrane assay. 2DG potentiated the cisplatin-induced inhibition of cell viability in a cisplatin-resistant subline. This study highlights HK2 as a prognostic biomarker for UBC patients and demonstrates the potential benefits of using 2DG as a glycolysis inhibitor. Future studies should focus on integrating 2DG into chemotherapy design, as an attempt to overcome cisplatin resistance.
Collapse
Affiliation(s)
- Julieta Afonso
- Life and Health Sciences Research Institute (ICVS), University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Céline Gonçalves
- Life and Health Sciences Research Institute (ICVS), University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Marta Costa
- Life and Health Sciences Research Institute (ICVS), University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Débora Ferreira
- Centre of Biological Engineering (CEB), University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
- LABBELS—Associate Laboratory, 4710-057 Braga, Portugal
| | - Lúcio Santos
- Experimental Pathology and Therapeutics Group, Research Center of the Portuguese Institute of Oncology (CI-IPOP), 4200-072 Porto, Portugal
- Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal
| | - Adhemar Longatto-Filho
- Life and Health Sciences Research Institute (ICVS), University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Laboratory of Medical Investigation (LIM14), Faculty of Medicine, São Paulo State University, São Paulo 01049-010, Brazil
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo 14784-400, Brazil
| | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS), University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Correspondence: ; Tel.: +351-253-60-48-28
| |
Collapse
|
5
|
Gerrits T, Brouwer IJ, Dijkstra KL, Wolterbeek R, Bruijn JA, Scharpfenecker M, Baelde HJ. Endoglin Is an Important Mediator in the Final Common Pathway of Chronic Kidney Disease to End-Stage Renal Disease. Int J Mol Sci 2022; 24:ijms24010646. [PMID: 36614087 PMCID: PMC9820946 DOI: 10.3390/ijms24010646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/15/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Chronic kidney disease (CKD) is a slow-developing, progressive deterioration of renal function. The final common pathway in the pathophysiology of CKD involves glomerular sclerosis, tubular atrophy and interstitial fibrosis. Transforming growth factor-beta (TGF-β) stimulates the differentiation of fibroblasts towards myofibroblasts and the production of extracellular matrix (ECM) molecules, and thereby interstitial fibrosis. It has been shown that endoglin (ENG, CD105), primarily expressed in endothelial cells and fibroblasts, can function as a co-receptor of TGF signaling. In several human organs, endoglin tends to be upregulated when chronic damage and fibrosis is present. We hypothesize that endoglin is upregulated in renal interstitial fibrosis and plays a role in the progression of CKD. We first measured renal endoglin expression in biopsy samples obtained from patients with different types of CKD, i.e., IgA nephropathy, focal segmental glomerulosclerosis (FSGS), diabetic nephropathy (DN) and patients with chronic allograft dysfunction (CAD). We showed that endoglin is upregulated in CAD patients (p < 0.001) and patients with DN (p < 0.05), compared to control kidneys. Furthermore, the amount of interstitial endoglin expression correlated with eGFR (p < 0.001) and the amount of interstitial fibrosis (p < 0.001), independent of the diagnosis of the biopsies. Finally, we investigated in vitro the effect of endoglin overexpression in TGF-β stimulated human kidney fibroblasts. Overexpression of endoglin resulted in an enhanced ACTA2, CCN2 and SERPINE1 mRNA response (p < 0.05). It also increased the mRNA and protein upregulation of the ECM components collagen type I (COL1A1) and fibronectin (FN1) (p < 0.05). Our results suggest that endoglin is an important mediator in the final common pathway of CKD and could be used as a possible new therapeutic target to counteract the progression towards end-stage renal disease (ESRD).
Collapse
Affiliation(s)
- Tessa Gerrits
- Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Isabella J. Brouwer
- Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Kyra L. Dijkstra
- Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Ron Wolterbeek
- Department of Biomedical Data Sciences, Medical Statistics, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Jan A. Bruijn
- Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Marion Scharpfenecker
- Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Hans J. Baelde
- Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
- Correspondence: ; Tel.: +31-(0)-71-526-4788
| |
Collapse
|
6
|
Moreno DA, da Silva LS, Gomes I, Leal LF, Berardinelli GN, Gonçalves GM, Pereira CA, Santana IVV, Matsushita MDM, Bhat K, Lawler S, Reis RM. Cancer immune profiling unveils biomarkers, immunological pathways, and cell type score associated with glioblastoma patients' survival. Ther Adv Med Oncol 2022; 14:17588359221127678. [PMID: 36579028 PMCID: PMC9791289 DOI: 10.1177/17588359221127678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/02/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Glioblastoma (GBM), isocitrate dehydrogenase (IDH) wild-type (IDH wt), and grade 4 astrocytomas, IDH mutant (IDH mut), are the most common and aggressive primary malignant brain tumors in adults. A better understanding of the tumor immune microenvironment may provide new biomarkers and therapeutic opportunities. Objectives We aimed to evaluate the expression profile of 730 immuno-oncology-related genes in patients with IDH wt GBM and IDH mut tumors and identify prognostic biomarkers and a gene signature associated with patient survival. Methods RNA was isolated from formalin-fixed, paraffin-embedded sections of 99 tumor specimens from patients treated with standard therapy. Gene expression profile was assessed using the Pan-Cancer Immune Profiling Panel (Nanostring Technologies, Inc., Seattle, WA, USA). Data analysis was performed using nSolverSoftware and validated in The Cancer Genome Atlas. In addition, we developed a prognostic signature using the cox regression algorithm (Least Absolute Shrinkage and Selection Operator). Results We found 88 upregulated genes, high immunological functions, and a high macrophage score in IDH wt GBM compared to IDH mut tumors. Regarding IDH wt GBM, we found 24 upregulated genes in short-term survivors (STS) and overexpression of CD274 (programmed death-ligand 1, PD-L1). Immune pathways, CD45, cytotoxic, and macrophage scores were upregulated in STS. Two different prognostic groups were found based on the 12-gene signature (CXCL14, PSEN2, TNFRSF13C, IL13RA1, MAP2K1, TNFSF14, THY1, CTSL, ITGAE, CHUK, CD207, and IFITM1). Conclusion The elevated expression of immune-oncology-related genes was associated with worse outcome in IDH wt GBM patients. Increased immune functions, CD45, cytotoxic cells, and macrophage scores were associated with a more aggressive phenotype and may provide promising possibilities for therapy. Moreover, a 12 gene-based signature could predict patients' prognosis.
Collapse
Affiliation(s)
| | | | - Isabella Gomes
- Molecular Oncology Research Center, Barretos, São Paulo, Brazil
| | | | | | | | | | | | | | - Krishna Bhat
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sean Lawler
- Harvard Medical School, Boston, MA, USA Brown University, Pathology and Laboratory Medicine, Providence, Rhode Island, USA
| | | |
Collapse
|
7
|
Construction of Prognostic Risk Model for Small Cell Lung Cancer Based on Immune-Related Genes. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7116080. [PMID: 36245844 PMCID: PMC9554662 DOI: 10.1155/2022/7116080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/30/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022]
Abstract
Small cell lung cancer (SCLC) is a highly invasive and fatal malignancy. Research at the present stage implied that the expression of immune-related genes is associated with the prognosis in SCLC. Accordingly, it is essential to explore effective immune-related molecular markers to judge prognosis and treat SCLC. Our research obtained SCLC dataset from Gene Expression Omnibus (GEO) and subjected mRNAs in it to differential expression analysis. Differentially expressed mRNAs (DEmRNAs) were intersected with immune-related genes to yield immune-related differentially expressed genes (DEGs). The functions of these DEGs were revealed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. Thereafter, we categorized 3 subtypes of immune-related DEGs via K-means clustering. Kaplan-Meier curves analyzed the effects of 3 subtypes on SCLC patients' survival. Single-sample gene set enrichment analysis (ssGSEA) and ESTIMATE validated that the activation of different immune gene subtypes differed significantly. Finally, an immune-related-7-gene assessment model was constructed by univariate-Lasso-multiple Cox regression analyses. Riskscores, Kaplan-Meier curves, receiver operating characteristic (ROC) curves, and independent prognostic analyses validated the prognostic value of the immune-related-7-gene assessment model. As suggested by GSEA, there was a prominent difference in cytokine-related pathways between high- and low-risk groups. As the analysis went further, we discovered a statistically significant difference in the expression of human leukocyte antigen (HLA) proteins and costimulatory molecules expressed on the surface of CD274, CD152, and T lymphocytes in different groups. In a word, we started with immune-related genes to construct the prognostic model for SCLC, which could effectively evaluate the clinical outcomes and offer guidance for the treatment and prognosis of SCLC patients.
Collapse
|
8
|
Li L, Zhong L, Tang C, Gan L, Mo T, Na J, He J, Huang Y. CD105: tumor diagnosis, prognostic marker and future tumor therapeutic target. Clin Transl Oncol 2022; 24:1447-1458. [PMID: 35165838 DOI: 10.1007/s12094-022-02792-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/21/2022] [Indexed: 02/06/2023]
Abstract
Cancer is one of the diseases with the highest morbidity and mortality rates worldwide, and its therapeutic options are inadequate. The endothelial glycoprotein, also known as CD105, is a type I transmembrane glycoprotein located on the surface of the cell membranes and it is one of the transforming growth factor-β (TGF-β) receptor complexes. It regulates the responses associated with binding to transforming growth factor β1 egg (Activin-A), bone morphogenetic protein 2 (BMP-2), and bone morphogenetic protein 7 (BMP-7). Additionally, it is involved in the regulation of angiogenesis. This glycoprotein is indispensable in the treatment of tumor angiogenesis, and it also plays a leading role in tumor angiogenesis therapy. Therefore, CD105 is considered to be a novel therapeutic target. In this study, we explored the significance of CD105 in the diagnosis, treatment and prognosis of various tumors, and provided evidence for the effect and mechanism of CD105 on tumors.
Collapse
Affiliation(s)
- Lan Li
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Liping Zhong
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chao Tang
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Lu Gan
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Tong Mo
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jintong Na
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jian He
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yong Huang
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
9
|
Galanis E, Anderson SK, Twohy E, Butowski NA, Hormigo A, Schiff D, Omuro A, Jaeckle KA, Kumar S, Kaufmann TJ, Geyer S, Kumthekar PU, Campian J, Giannini C, Buckner JC, Wen PY. Phase I/randomized phase II trial of TRC105 plus bevacizumab versus bevacizumab in recurrent glioblastoma: North Central Cancer Treatment Group N1174 (Alliance). Neurooncol Adv 2022; 4:vdac041. [PMID: 35664553 PMCID: PMC9154335 DOI: 10.1093/noajnl/vdac041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Patients with glioblastoma (GBM) have a poor prognosis and limited effective treatment options. Bevacizumab has been approved for treatment of recurrent GBM, but there is questionable survival benefit. Based on preclinical and early clinical data indicating that CD105 upregulation may represent a mechanism of resistance to bevacizumab, we hypothesized that combining bevacizumab with the anti-CD105 antibody TRC105 may improve efficacy in recurrent GBM. Methods Phase I dose-escalation/comparative randomized phase II trial in patients with GBM. During phase I, the maximum tolerated dose (MTD) of TRC105 in combination with bevacizumab was determined. In phase II, patients were randomized 1:1 to TRC105 and bevacizumab or bevacizumab monotherapy. Patients received TRC105 (10 mg/kg) weekly and bevacizumab (10 mg/kg) every 2 weeks. Efficacy, as assessed by progression-free survival (PFS), was the primary endpoint; safety, quality of life, and correlative outcomes were also evaluated. Results In total, 15 patients were enrolled in phase I and 101 in phase II; 52 patients were randomized to TRC105 with bevacizumab and 49 to bevacizumab monotherapy. The MTD was determined to be 10 mg/kg TRC105 weekly plus bevacizumab 10 mg/kg every 2 weeks. An increased occurrence of grade ≥3 adverse events was seen in the combination arm, including higher incidences of anemia. Median PFS was similar in both treatment arms: 2.9 months for combination versus 3.2 months for bevacizumab monotherapy (HR = 1.16, 95% CI = 0.75-1.78, P = .51). Quality of life scores were similar for both treatment arms. Conclusions TRC105 in combination with bevacizumab was well tolerated in patients with recurrent GBM, but no difference in efficacy was observed compared to bevacizumab monotherapy.
Collapse
Affiliation(s)
- Evanthia Galanis
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Pathology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo Clinic, Rochester, Minnesota, USA
| | - S Keith Anderson
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Erin Twohy
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Nicholas A Butowski
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Adilia Hormigo
- Department of Neurology, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, New York, USA
| | - David Schiff
- Department of Neurology, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Antonio Omuro
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kurt A Jaeckle
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Shaji Kumar
- Department of Hematology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo Clinic, Rochester, Minnesota, USA
| | - Timothy J Kaufmann
- Department of Neuroradiology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo Clinic, Rochester, Minnesota, USA
| | - Susan Geyer
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Priya U Kumthekar
- Northwestern Medicine, Lou and Jean Malnati Brain Tumor Institute, Chicago Illinois, USA
| | - Jian Campian
- Washington University School of Medicine, Siteman Cancer Center, St. Louis, Missouri, USA
| | | | - Jan C Buckner
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo Clinic, Rochester, Minnesota, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Partners Cancer Care, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Minhajat R, Saleh S, Harjianti T, Benyamin AF, Parewangi AML, Bakri S. Profiles of Endoglin and Vascular Endothelial Growth Factor Based on Staging and Histological Grading of Colorectal Cancer and Their Relationship with Bevacizumab Therapy. JOURNAL OF COLOPROCTOLOGY 2021. [DOI: 10.1055/s-0041-1724065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Abstract
Objective The present study evaluated the profile of endoglin (CD105) and vascular endothelial growth factor (VEGF) based on staging and histopathological grading of colorectal cancer as well as their relationship with bevacizumab therapy.
Methods A total of 88 cases of colorectal adenocarcinoma were included in the present study. The levels of VEGF and CD105 protein were evaluated with enzyme-linked immunosorbent assay (ELISA).
Results There was a significant difference in the level of CD105 (p = 0.002) between metastases and non-metastases subjects, showing that CD105 was higher in metastases subjects (4.59 ng/ml). There was no significant difference in the level of VEGF based on the presence of metastasis (p = 0.625). There was a significant difference in the levels of CD105 (p = 0.038) and VEGF (p = 0.010) between the subjects who received chemotherapy and those who did not. The CD105 level was higher in the subjects who received chemotherapy (4.43 ng/ml); conversely, the level of VEGF was lower in subjects who received chemotherapy (543.65 pg/ml). There was a statistically significant difference in the levels of CD105 (p = 0.003) and VEGF (p = 0.002) between subjects who received bevacizumab therapy and subjects who did not. The levels of CD105 were higher in subjects who received bevacizumab therapy (5.11 ng/ml); in contrast, the level of VEGF was higher in subjects who did not receive bevacizumab therapy (645.92 pg/ml). There was a significant positive correlation between CD105 and VEGF in subjects who did not receive bevacizumab (p < 0.01).
Conclusion The results of this study support a hypothesis of “escape mechanism” in the failure of anti-angiogenesis therapy (anti-VEGF).
Collapse
Affiliation(s)
- Rahmawati Minhajat
- Division of Hematology and Medical Oncology, Internal Medicine Department, Faculty of Medicine at Hasanuddin University, Makassar, Indonesia
- Department of Histology, Faculty of Medicine at Hasanuddin University, Makassar, Indonesia
| | - Sahyuddin Saleh
- Division of Hematology and Medical Oncology, Internal Medicine Department, Faculty of Medicine at Hasanuddin University, Makassar, Indonesia
| | - Tutik Harjianti
- Division of Hematology and Medical Oncology, Internal Medicine Department, Faculty of Medicine at Hasanuddin University, Makassar, Indonesia
| | - Andi Fachruddin Benyamin
- Division of Hematology and Medical Oncology, Internal Medicine Department, Faculty of Medicine at Hasanuddin University, Makassar, Indonesia
| | - A. M. Luthfi Parewangi
- Division of Gastroenterology and Hepatology, Internal Medicine Department, Faculty of Medicine at Hasanuddin University, Makassar, Indonesia
| | - Syakib Bakri
- Division of Nephrology and Hypertension, Internal Medicine Department, Faculty of Medicine at Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
11
|
Choueiri TK, Zakharia Y, Pal S, Kocsis J, Pachynski R, Poprach A, Nixon AB, Liu Y, Starr M, Lyu J, Owzar K, deShazo M, Lara P, Geczi L, Ho TH, Walsh M, Adams B, Robertson L, Darif M, Theuer C, Agarwal N. Clinical Results and Biomarker Analyses of Axitinib and TRC105 versus Axitinib Alone in Patients with Advanced or Metastatic Renal Cell Carcinoma (TRAXAR). Oncologist 2021; 26:560-e1103. [PMID: 33829609 DOI: 10.1002/onco.13777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/05/2021] [Indexed: 12/17/2022] Open
Abstract
LESSONS LEARNED The combination of carotuximab with axitinib did not provide a benefit over axitinib monotherapy in patients with metastatic clear cell renal cell carcinoma who had previously progressed on one or more vascular endothelial growth factor (VEGF)-targeted therapies. Exploratory evaluation of pretreatment circulating biomarkers suggested the combination might benefit patients who have low baseline VEGF levels. BACKGROUND Endoglin is an angiogenic receptor expressed on proliferating tumor vessels and renal cell carcinoma (RCC) stem cells that is implicated as a mechanism of resistance to vascular endothelial growth factor receptor (VEGFR) inhibitors. This study evaluated an antiendoglin monoclonal antibody (carotuximab, TRC105) combined with axitinib in patients with advanced or metastatic clear cell renal cell carcinoma (mccRCC) who had progressed following one or more prior VEGF inhibitors. METHODS TRAXAR was a multicenter, international randomized 1:1 (stratified by ECOG, 0 vs. 1), phase II study of carotuximab combined with axitinib versus axitinib alone in mccRCC patients who had progressed following one or more prior VEGF inhibitors. The primary endpoint was progression-free survival (PFS) assessed by independent central review (ICR) per RECIST 1.1 RESULTS: A total of 150 patients were randomized. The combination therapy resulted in shorter median PFS by RECIST 1.1 than axitinib monotherapy (6.7 vs. 11.4 months). The combination was tolerated similarly to axitinib monotherapy, and there were no treatment related deaths. Exploratory evaluation of pretreatment circulating biomarkers suggested the combination might benefit patients who have low baseline VEGF levels. CONCLUSION The combination of carotuximab with axitinib did not demonstrate additional efficacy over single agent axitinib in patients with mccRCC who progressed following one or more prior VEGF inhibitor treatment.
Collapse
Affiliation(s)
| | - Yousef Zakharia
- University of Iowa, Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
| | - Sumanta Pal
- City of Hope National Medical Center, Duarte, California, USA
| | - Judit Kocsis
- Bács-Kiskun County Hospital, Oncoradiology Center, Kecskemét, Hungary
| | - Russell Pachynski
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alexandr Poprach
- Department of Comprehensive Cancer Care and Faculty of Medicine, Masaryk Memorial Cancer Institute and Masaryk University, Brno, Czech Republic
| | - Andrew B Nixon
- Department of Medicine, Duke University Medical Center, Durham, North, Carolina, USA
| | - Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, North, Carolina, USA
| | - Mark Starr
- Department of Medicine, Duke University Medical Center, Durham, North, Carolina, USA
| | - Jing Lyu
- Graduate Group in Biostatistics, University of California Davis, Davis, California, USA
| | - Kouros Owzar
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, USA
| | - Mollie deShazo
- Division of Hematology/Oncology, Department of Internal Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Primo Lara
- University of California, Davis Medical Center, Sacramento, California, USA
| | - Lajos Geczi
- Országos Onkológiai Intézet, Budapest, Hungary
| | - Thai H Ho
- Division of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Meghara Walsh
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Bonne Adams
- TRACON Pharmaceuticals, Inc., San Diego, California, USA
| | - Liz Robertson
- TRACON Pharmaceuticals, Inc., San Diego, California, USA
| | - Mohamed Darif
- TRACON Pharmaceuticals, Inc., San Diego, California, USA
| | - Charles Theuer
- TRACON Pharmaceuticals, Inc., San Diego, California, USA
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
12
|
Zhong L, Shi W, Gan L, Liu X, Huo Y, Wu P, Zhang Z, Wu T, Peng H, Huang Y, Zhao Y, Yuan Y, Deng Z, Tang H. Human endoglin-CD3 bispecific T cell engager antibody induces anti-tumor effect in vivo. Am J Cancer Res 2021; 11:6393-6406. [PMID: 33995664 PMCID: PMC8120215 DOI: 10.7150/thno.53121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 03/12/2021] [Indexed: 12/11/2022] Open
Abstract
Rationale: Endoglin, also known as CD105, is a homo-dimeric membrane glycoprotein required for angiogenesis and serves as a marker for cancer vasculature. In this study, we constructed a bispecific T-cell engager (BiTE) antibody that targets human endoglin and CD3 (hEND-CD3/BiTE). We examined BiTE binding to endoglin-expressing cells and its effects on the cytolytic activity of T cells and cancer development. Methods: The in vitro effects of hEND-CD3/BiTE, including binding to target cells, T-cell activation, proliferation, and cytotoxicity, were examined in endoglin-expressing 293T cells, human umbilical vascular endothelial cells, tumor-derived endothelial cells, and CD3+ T cells. An in vivo xenograft tumor model was established using A549 human lung cancer cells. The therapeutic efficacy of hEND-CD3/BiTE was assessed by monitoring tumor growth, angiogenesis, and mouse survival. Results: hEND-CD3/BiTE specifically bound to endoglin-expressing cells and CD3+ T cells in vitro and stimulated T-cell activation, proliferation, and Th1 cytokine secretion, and promoted T-cell-mediated cytolysis of endoglin-expressing cells. The hEND-CD3/BiTE in vivo caused minimal toxicity to major organs, reduced tumor neoangiogenesis, inhibited tumor growth, and significantly improved mouse survival. Conclusions: Our study demonstrated the therapeutic potential of hEND-CD3/BiTE and provided a novel approach to clinical cancer treatment.
Collapse
|
13
|
Ahmed T, Mythreye K, Lee NY. Strength and duration of GIPC-dependent signaling networks as determinants in cancer. Neoplasia 2021; 23:181-188. [PMID: 33360508 PMCID: PMC7773760 DOI: 10.1016/j.neo.2020.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 10/25/2022]
Abstract
GIPC is a PDZ-domain containing adaptor protein that regulates the cell surface expression and endocytic trafficking of numerous transmembrane receptors and signaling complexes. Interactions with over 50 proteins have been reported to date including VEGFR, insulin-like growth factor-1 receptor (IGF-1R), GPCRs, and APPL, many of which have essential roles in neuronal and cardiovascular development. In cancer, a major subset of GIPC-binding receptors and cytoplasmic effectors have been shown to promote tumorigenesis or metastatic progression, while other subsets have demonstrated strong tumor-suppressive effects. Given that these diverse pathways are widespread in normal tissues and human malignancies, precisely how these opposing signals are integrated and regulated within the same tumor setting likely depend on the strength and duration of their interactions with GIPC. This review highlights the major pathways and divergent mechanisms of GIPC signaling in various cancers and provide a rationale for emerging GIPC-targeted cancer therapies.
Collapse
Affiliation(s)
- Tasmia Ahmed
- Deparment of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Karthikeyan Mythreye
- Division of Molecular and Cellular Pathology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Nam Y Lee
- Deparment of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA; Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, Tucson, AZ, USA.
| |
Collapse
|
14
|
Liu Y, Paauwe M, Nixon AB, Hawinkels LJ. Endoglin Targeting: Lessons Learned and Questions That Remain. Int J Mol Sci 2020; 22:ijms22010147. [PMID: 33375670 PMCID: PMC7795616 DOI: 10.3390/ijms22010147] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Approximately 30 years ago, endoglin was identified as a transforming growth factor (TGF)-β coreceptor with a crucial role in developmental biology and tumor angiogenesis. Its selectively high expression on tumor vessels and its correlation with poor survival in cancer patients led to the exploration of endoglin as a therapeutic target for cancer. The endoglin neutralizing antibody TRC105 (Carotuximab®, Tracon Pharmaceuticals (San Diego, CA, USA) was subsequently tested in a wide variety of preclinical cancer models before being tested in phase I-III clinical studies in cancer patients as both a monotherapy and in combination with other chemotherapeutic and anti-angiogenic therapies. The combined data of these studies have revealed new insights into the role of endoglin in angiogenesis and its expression and functional role on other cells in the tumor microenvironment. In this review, we will summarize the preclinical work, clinical trials and biomarker studies of TRC105 and explore what these studies have enabled us to learn and what questions remain unanswered.
Collapse
Affiliation(s)
- Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; (Y.L.); (A.B.N.)
| | - Madelon Paauwe
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Andrew B. Nixon
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; (Y.L.); (A.B.N.)
| | - Lukas J.A.C. Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Correspondence: ; Tel.: +31-71-526-6736
| |
Collapse
|
15
|
Endoglin: An 'Accessory' Receptor Regulating Blood Cell Development and Inflammation. Int J Mol Sci 2020; 21:ijms21239247. [PMID: 33287465 PMCID: PMC7729465 DOI: 10.3390/ijms21239247] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1) is a pleiotropic factor sensed by most cells. It regulates a broad spectrum of cellular responses including hematopoiesis. In order to process TGF-β1-responses in time and space in an appropriate manner, there is a tight regulation of its signaling at diverse steps. The downstream signaling is mediated by type I and type II receptors and modulated by the ‘accessory’ receptor Endoglin also termed cluster of differentiation 105 (CD105). Endoglin was initially identified on pre-B leukemia cells but has received most attention due to its high expression on activated endothelial cells. In turn, Endoglin has been figured out as the causative factor for diseases associated with vascular dysfunction like hereditary hemorrhagic telangiectasia-1 (HHT-1), pre-eclampsia, and intrauterine growth restriction (IUPR). Because HHT patients often show signs of inflammation at vascular lesions, and loss of Endoglin in the myeloid lineage leads to spontaneous inflammation, it is speculated that Endoglin impacts inflammatory processes. In line, Endoglin is expressed on progenitor/precursor cells during hematopoiesis as well as on mature, differentiated cells of the innate and adaptive immune system. However, so far only pro-monocytes and macrophages have been in the focus of research, although Endoglin has been identified in many other immune system cell subsets. These findings imply a functional role of Endoglin in the maturation and function of immune cells. Aside the functional relevance of Endoglin in endothelial cells, CD105 is differentially expressed during hematopoiesis, arguing for a role of this receptor in the development of individual cell lineages. In addition, Endoglin expression is present on mature immune cells of the innate (i.e., macrophages and mast cells) and the adaptive (i.e., T-cells) immune system, further suggesting Endoglin as a factor that shapes immune responses. In this review, we summarize current knowledge on Endoglin expression and function in hematopoietic precursors and mature hematopoietic cells of different lineages.
Collapse
|
16
|
Baik J, Felices M, Yingst A, Theuer CP, Verneris MR, Miller JS, Perlingeiro R. Therapeutic effect of TRC105 and decitabine combination in AML xenografts. Heliyon 2020; 6:e05242. [PMID: 33088975 PMCID: PMC7566100 DOI: 10.1016/j.heliyon.2020.e05242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/30/2020] [Accepted: 10/08/2020] [Indexed: 01/13/2023] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy, often characterized by poor prognosis following standard induction therapy. The hypomethylating agent decitabine (DAC) is an alternative treatment for elderly and relapsed/refractory AML patients, yet responses following DAC monotherapy are still modest. The transforming growth factor-β (TGF-β) receptor CD105 (endoglin) is expressed in various hematopoietic malignancies, and high CD105 expression correlates with poor prognosis in AML patients. Using a xenograft model, we have recently demonstrated that targeting CD105+ AML blasts with the TRC105 monoclonal antibody inhibits leukemia progression. Here we investigated whether administration of TRC105 along with DAC could represent a novel therapeutic option for relapsed/refractory AML. Our data show that the DAC/TRC105 combination results in a more durable anti-leukemic effect in AML xenografts compared to DAC monotherapy. Moreover, the DAC/TRC105 combination enhanced reactive oxygen species (ROS) activity, which correlated with reduced leukemia burden. RNA-sequencing studies suggest that TRC105 may alter TGF-β activity in AML blasts. Taken together, these findings provide rationale for the clinical evaluation of TRC105 in combination with DAC in AML patients.
Collapse
Affiliation(s)
- June Baik
- Dept. of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Martin Felices
- Dept. of Medicine, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Ashley Yingst
- Dept. of Pediatrics, University of Denver, Colorado, CO, USA
| | | | | | - Jeffrey S Miller
- Dept. of Medicine, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Rita Perlingeiro
- Dept. of Medicine, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
17
|
Upregulation of annexin A1 protein expression in the intratumoral vasculature of human non-small-cell lung carcinoma and rodent tumor models. PLoS One 2020; 15:e0234268. [PMID: 32497150 PMCID: PMC7272081 DOI: 10.1371/journal.pone.0234268] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/21/2020] [Indexed: 12/23/2022] Open
Abstract
Annexin A1 (anxA1) is an immunomodulatory protein that has been proposed as a tumor vascular target for antitumor biologic agents, yet to date the vascular expression of anxA1 in specific tumor indications has not been systematically assessed. Attempts to evaluate vascular anxA1 expression by immunohistochemistry are complicated by a lack of available antibodies that are both specific for anxA1 and bind the N-terminal–truncated form of anxA1 that has previously been identified in tumor vasculature. To study the vascular expression pattern of anxA1 in non–small-cell lung carcinoma (NSCLC), we isolated an antibody capable of binding N-terminal–truncated anxA127-346 and employed it in immunohistochemical studies of human lung specimens. Lung tumor specimens evaluated with this antibody revealed vascular (endothelial) anxA1 expression in five of eight tumor samples studied, but no vascular anxA1 expression was observed in normal lung tissue. Tumor microarray analysis further demonstrated positive vascular staining for anxA1 in 30 of 80 NSCLC samples, and positive staining of neoplastic cells was observed in 54 of 80 samples. No correlation was observed between vascular and parenchymal anxA1 expression. Two rodent tumor models, B16-F10 and Py230, were determined to have upregulated anxA1 expression in the intratumoral vasculature. These data validate anxA1 as a potential vascular anti-tumor target in a subset of human lung tumors and identify rodent models which demonstrate anxA1 expression in tumor vasculature.
Collapse
|
18
|
Schoonderwoerd MJA, Koops MFM, Angela RA, Koolmoes B, Toitou M, Paauwe M, Barnhoorn MC, Liu Y, Sier CFM, Hardwick JCH, Nixon AB, Theuer CP, Fransen MF, Hawinkels LJAC. Targeting Endoglin-Expressing Regulatory T Cells in the Tumor Microenvironment Enhances the Effect of PD1 Checkpoint Inhibitor Immunotherapy. Clin Cancer Res 2020; 26:3831-3842. [PMID: 32332012 DOI: 10.1158/1078-0432.ccr-19-2889] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 03/16/2020] [Accepted: 04/21/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Endoglin is a coreceptor for TGFβ ligands that is highly expressed on proliferating endothelial cells and other cells in the tumor microenvironment. Clinical studies have noted increased programmed cell death (PD)-1 expression on cytotoxic T cells in the peripheral blood of patients with cancer treated with TRC105, an endoglin-targeting antibody. In this study, we investigated the combination of endoglin antibodies (TRC105 and M1043) with an anti-PD1 antibody. EXPERIMENTAL DESIGN The combination anti-endoglin/anti-PD1 antibodies was tested in four preclinical mouse models representing different stages of cancer development. To investigate the underlying mechanism, Fc-receptor-knockout mice were used complemented with depletion of multiple immune subsets in mice. Tumor growth and the composition of immune infiltrate were analyzed by flow cytometry. Finally, human colorectal cancer specimens were analyzed for presence of endoglin-expressing regulatory T cells (Treg). RESULTS In all models, the combination of endoglin antibody and PD1 inhibition produced durable tumor responses, leading to complete regressions in 30% to 40% of the mice. These effects were dependent on the presence of Fcγ receptors, indicating the involvement of antibody-dependent cytotoxic responses and the presence of CD8+ cytotoxic T cells and CD4+ Th cells. Interestingly, treatment with the endoglin antibody, TRC105, significantly decreased the number of intratumoral Tregs. Endoglin-expressing Tregs were also detected in human colorectal cancer specimens. CONCLUSIONS Taken together, these data provide a rationale for combining TRC105 and anti-PD1 therapy and provide additional evidence of endoglin's immunomodulatory role.
Collapse
Affiliation(s)
- Mark J A Schoonderwoerd
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Maaike F M Koops
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ricardo A Angela
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Bryan Koolmoes
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Melpomeni Toitou
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Madelon Paauwe
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke C Barnhoorn
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - James C H Hardwick
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Andrew B Nixon
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | | | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Lukas J A C Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
19
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
20
|
Schoonderwoerd MJA, Goumans MJTH, Hawinkels LJAC. Endoglin: Beyond the Endothelium. Biomolecules 2020; 10:biom10020289. [PMID: 32059544 PMCID: PMC7072477 DOI: 10.3390/biom10020289] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 02/06/2023] Open
Abstract
Keywords: endoglin; CD105 TGF-β; BMP9; ALK-1; TRC105; tumor microenvironment.
Collapse
Affiliation(s)
- Mark J. A. Schoonderwoerd
- Department of Gastrenterology-Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | - Lukas J. A. C. Hawinkels
- Department of Gastrenterology-Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- Correspondence: ; Tel.: +31-71-526-6736
| |
Collapse
|
21
|
Egorova AA, Shtykalova SV, Maretina MA, Sokolov DI, Selkov SA, Baranov VS, Kiselev AV. Synergistic Anti-Angiogenic Effects Using Peptide-Based Combinatorial Delivery of siRNAs Targeting VEGFA, VEGFR1, and Endoglin Genes. Pharmaceutics 2019; 11:E261. [PMID: 31174285 PMCID: PMC6631635 DOI: 10.3390/pharmaceutics11060261] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 12/18/2022] Open
Abstract
Angiogenesis is a process of new blood vessel formation, which plays a significant role in carcinogenesis and the development of diseases associated with pathological neovascularization. An important role in the regulation of angiogenesis belongs to several key pathways such as VEGF-pathways, TGF-β-pathways, and some others. Introduction of small interfering RNA (siRNA) against genes of pro-angogenic factors is a promising strategy for the therapeutic suppression of angiogenesis. These siRNA molecules need to be specifically delivered into endothelial cells, and non-viral carriers modified with cellular receptor ligands can be proposed as perspective delivery systems for anti-angiogenic therapy purposes. Here we used modular peptide carrier L1, containing a ligand for the CXCR4 receptor, for the delivery of siRNAs targeting expression of VEGFA, VEGFR1 and endoglin genes. Transfection properties of siRNA/L1 polyplexes were studied in CXCR4-positive breast cancer cells MDA-MB-231 and endothelial cells EA.Hy926. We have demonstrated the efficient down-regulation of endothelial cells migration and proliferation by anti-VEGFA, anti-VEGFR1, and anti-endoglin siRNA-induced silencing. It was found that the efficiency of anti-angiogenic treatment can be synergistically improved via the combinatorial delivery of anti-VEGFA and anti-VEGFR1 siRNAs. Thus, this approach can be useful for the development of therapeutic angiogenesis inhibition.
Collapse
Affiliation(s)
- Anna A Egorova
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
| | - Sofia V Shtykalova
- Department of Genetics and Biotechnology, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia.
| | - Marianna A Maretina
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
| | - Dmitry I Sokolov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
| | - Sergei A Selkov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
| | - Vladislav S Baranov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
- Department of Genetics and Biotechnology, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia.
| | - Anton V Kiselev
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
| |
Collapse
|
22
|
Chen W, Dong W, Wang J, Wen Z, Hao X. Elevated Expressions of Survivin and Endoglin in Patients with Hepatic Carcinoma. Cancer Biother Radiopharm 2019; 34:7-12. [PMID: 30730204 DOI: 10.1089/cbr.2018.2539] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE To investigate expression profiles of survivin and endoglin in patients with hepatic carcinoma. MATERIALS AND METHODS Cancerous tissues (hepatic carcinoma group) of 48 patients with hepatic carcinoma and adjacent noncancerous hepatic tissues (control group) were used as objects of study. Histopathological staining [hematoxylin & eosin (H&E) staining] was used to study the pathological differences in hepatic tissues between hepatic carcinoma group and control group. Moreover, survivin and endoglin protein expressions in hepatic tissues in hepatic carcinoma group and control group were detected via western blotting. Finally, Statistical Product and Service Solutions 17.0 statistical software was used to analyze the differences in survivin and endoglin expressions in hepatic tissues between hepatic carcinoma group and control group. RESULTS H&E staining showed that histopathological features in hepatic carcinoma group were significantly different from those in control group. Compared with those in control group, the cell structure in hepatic carcinoma group was damaged, karyopyknosis was obvious, and the hepatic injury was serious. Reverse transcription-polymerase chain reaction showed that survivin and endoglin mRNA expression levels in hepatic carcinoma group were significantly increased compared with those in control group. Besides, immunofluorescence method and western blotting revealed the low expressions of survivin and endoglin proteins in tissues in control group, which were obviously lower than those in hepatic tissues in hepatic carcinoma group. Results of analyses of variance showed that the expressions of survivin and endoglin in normal hepatic tissues and cancerous tissues had statistically significant differences (p < 0.01). Furthermore, expressions of survivin and endoglin were significantly associated with histological grade, tumor size, and tumor, node, metastasis (TNM) stage. CONCLUSION Elevated expressions of survivin and endoglin are associated with histological grade, tumor size, and TNM stage in patients with hepatic carcinoma, indicating that survivin and endoglin might be involved in the pathogenesis of hepatic carcinoma and therapeutic targeting them might be a novel approach for the treatment of hepatic carcinoma.
Collapse
Affiliation(s)
- Wenmei Chen
- 1 Department of Liver Disease Area 9, Qingdao No.6 People's Hospital, Qingdao, China
| | - Wenjing Dong
- 2 Department of Liver Disease Area 7, Qingdao No.6 People's Hospital, Qingdao, China
| | - Jintai Wang
- 3 Department of Orthopedic, Qingdao No.8 People's Hospital, Qingdao, China
| | - Zirong Wen
- 1 Department of Liver Disease Area 9, Qingdao No.6 People's Hospital, Qingdao, China
| | - Xinjie Hao
- 2 Department of Liver Disease Area 7, Qingdao No.6 People's Hospital, Qingdao, China
| |
Collapse
|
23
|
Wu HW, Sheard MA, Malvar J, Fernandez GE, DeClerck YA, Blavier L, Shimada H, Theuer CP, Sposto R, Seeger RC. Anti-CD105 Antibody Eliminates Tumor Microenvironment Cells and Enhances Anti-GD2 Antibody Immunotherapy of Neuroblastoma with Activated Natural Killer Cells. Clin Cancer Res 2019; 25:4761-4774. [PMID: 31068371 DOI: 10.1158/1078-0432.ccr-18-3358] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/29/2019] [Accepted: 04/26/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE We determined whether elimination of CD105+ cells in the tumor microenvironment (TME) with anti-CD105 antibodies enhanced anti-disialoganglioside (GD2) antibody dinutuximab therapy of neuroblastoma when combined with activated natural killer (aNK) cells. EXPERIMENTAL DESIGN The effect of MSCs and monocytes on antibody-dependent cellular cytotoxicity (ADCC) mediated by dinutuximab with aNK cells against neuroblastoma cells was determined in vitro. ADCC with anti-CD105 mAb TRC105 and aNK cells against MSCs, monocytes, and endothelial cells, which express CD105, was evaluated. Anti-neuroblastoma activity in immunodeficient NSG mice of dinutuximab with aNK cells without or with anti-CD105 mAbs was determined using neuroblastoma cell lines and a patient-derived xenograft. RESULTS ADCC mediated by dinutuximab with aNK cells against neuroblastoma cells in vitro was suppressed by addition of MSCs and monocytes, and dinutuximab with aNK cells was less effective against neuroblastomas formed with coinjected MSCs and monocytes in NSG mice than against those formed by tumor cells alone. Anti-CD105 antibody TRC105 with aNK cells mediated ADCC against MSCs, monocytes, and endothelial cells. Neuroblastomas formed in NSG mice by two neuroblastoma cell lines or a patient-derived xenograft coinjected with MSCs and monocytes were most effectively treated with dinutuximab and aNK cells when anti-human (TRC105) and anti-mouse (M1043) CD105 antibodies were added, which depleted human MSCs and murine endothelial cells and macrophages from the TME. CONCLUSIONS Immunotherapy of neuroblastoma with anti-GD2 antibody dinutuximab and aNK cells is suppressed by CD105+ cells in the TME, but suppression is overcome by adding anti-CD105 antibodies to eliminate CD105+ cells.
Collapse
Affiliation(s)
- Hong-Wei Wu
- Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, California
| | - Michael A Sheard
- Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, California
| | - Jemily Malvar
- Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, California
| | - G Esteban Fernandez
- Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, California
| | - Yves A DeClerck
- Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, California.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Laurence Blavier
- Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, California
| | - Hiroyuki Shimada
- Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, California.,Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - Richard Sposto
- Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, California.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Robert C Seeger
- Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, California. .,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
24
|
Attia MF, Anton N, Wallyn J, Omran Z, Vandamme TF. An overview of active and passive targeting strategies to improve the nanocarriers efficiency to tumour sites. ACTA ACUST UNITED AC 2019; 71:1185-1198. [PMID: 31049986 DOI: 10.1111/jphp.13098] [Citation(s) in RCA: 527] [Impact Index Per Article: 87.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 04/07/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVES This review highlights both the physicochemical characteristics of the nanocarriers (NCs) and the physiological features of tumour microenvironment (TME) to outline what strategies undertaken to deliver the molecules of interest specifically to certain lesions. This review discusses these properties describing the convenient choice between passive and active targeting mechanisms with details, illustrated with examples of targeting agents up to preclinical research or clinical advances. KEY FINDINGS Targeted delivery approaches for anticancers have shown a steep rise over the past few decades. Though many successful preclinical trials, only few passive targeted nanocarriers are approved for clinical use and none of the active targeted nanoparticles. Herein, we review the principles and for both processes and the correlation with the tumour microenvironment. We also focus on the limitation and advantages of each systems regarding laboratory and industrial scale. SUMMARY The current literature discusses how the NCs and the enhanced permeation and retention effect impact the passive targeting. Whereas the active targeting relies on the ligand-receptor binding, which improves selective accumulation to targeted sites and thus discriminates between the diseased and healthy tissues. The latter could be achieved by targeting the endothelial cells, tumour cells, the acidic environment of cancers and nucleus.
Collapse
Affiliation(s)
- Mohamed F Attia
- CNRS, CAMB, UMR 7199, Université de Strasbourg, Strasbourg, France.,Department of Bioengineering, Clemson University, Clemson, SC, USA.,National Research Centre, Cairo, Egypt
| | - Nicolas Anton
- CNRS, CAMB, UMR 7199, Université de Strasbourg, Strasbourg, France
| | - Justine Wallyn
- CNRS, CAMB, UMR 7199, Université de Strasbourg, Strasbourg, France
| | - Ziad Omran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Umm Al-Qura, Kingdom of Saudi Arabia
| | | |
Collapse
|
25
|
Kasper B. The challenge of finding new therapeutic avenues in soft tissue sarcomas. Clin Sarcoma Res 2019; 9:5. [PMID: 31007895 PMCID: PMC6457009 DOI: 10.1186/s13569-019-0115-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/05/2019] [Indexed: 11/10/2022] Open
Abstract
Soft tissue sarcomas are rare malignancies of mesenchymal origin comprising about 1% of all adult cancers. Systemic therapies for locally advanced and metastatic disease have been restricted for decades to very few effective and approved agents such as doxorubicin and ifosfamide. However, new therapeutic avenues including new drug developments and registrations such as trabectedin, pazopanib and eribulin as well as numerous clinical trial options have recently enriched the therapeutic armamentarium in the treatment of patients with advanced soft tissue sarcomas. The challenges and pitfalls of finding such new therapeutic avenues in recent years for the treatment benefit of patients with soft tissue sarcomas will be presented in this chapter within the thematic series on "Challenges in Sarcoma".
Collapse
Affiliation(s)
- Bernd Kasper
- Sarcoma Unit, Interdisciplinary Tumor Center Mannheim, Mannheim University Medical Center, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| |
Collapse
|
26
|
Le BT, Raguraman P, Kosbar TR, Fletcher S, Wilton SD, Veedu RN. Antisense Oligonucleotides Targeting Angiogenic Factors as Potential Cancer Therapeutics. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 14:142-157. [PMID: 30594893 PMCID: PMC6307321 DOI: 10.1016/j.omtn.2018.11.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
Abstract
Cancer is one of the leading causes of death worldwide, and conventional cancer therapies such as surgery, chemotherapy, and radiotherapy do not address the underlying molecular pathologies, leading to inadequate treatment and tumor recurrence. Angiogenic factors, such as EGF, PDGF, bFGF, TGF-β, TGF-α, VEGF, endoglin, and angiopoietins, play important roles in regulating tumor development and metastasis, and they serve as potential targets for developing cancer therapeutics. Nucleic acid-based therapeutic strategies have received significant attention in the last two decades, and antisense oligonucleotide-mediated intervention is a prominent therapeutic approach for targeted manipulation of gene expression. Clinical benefits of antisense oligonucleotides have been recognized by the U.S. Food and Drug Administration, with full or conditional approval of Vitravene, Kynamro, Exondys51, and Spinraza. Herein we review the scope of antisense oligonucleotides that target angiogenic factors toward tackling solid cancers.
Collapse
Affiliation(s)
- Bao T Le
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Prithi Raguraman
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Tamer R Kosbar
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Susan Fletcher
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Steve D Wilton
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Rakesh N Veedu
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia.
| |
Collapse
|
27
|
Kasprzak A, Adamek A. Role of Endoglin (CD105) in the Progression of Hepatocellular Carcinoma and Anti-Angiogenic Therapy. Int J Mol Sci 2018; 19:E3887. [PMID: 30563158 PMCID: PMC6321450 DOI: 10.3390/ijms19123887] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/02/2018] [Accepted: 12/03/2018] [Indexed: 02/08/2023] Open
Abstract
The liver is perfused by both arterial and venous blood, with a resulting abnormal microenvironment selecting for more-aggressive malignancies. Hepatocellular carcinoma (HCC) is the most frequent primary liver cancer, the sixth most common cancer globally, and the third leading cause of cancer-related mortality worldwide. HCC is characterized by its hypervascularization. Improving the efficiency of anti-angiogenic treatment and mitigation of anti-angiogenic drug resistance are the top priorities in the development of non-surgical HCC therapies. Endoglin (CD105), a transmembrane glycoprotein, is one of the transforming growth factor β (TGF-β) co-receptors. Involvement of that protein in angiogenesis of solid tumours is well documented. Endoglin is a marker of activated endothelial cells (ECs), and is preferentially expressed in the angiogenic endothelium of solid tumours, including HCC. HCC is associated with changes in CD105-positive ECs within and around the tumour. The large spectrum of endoglin effects in the liver is cell-type- and HCC- stage-specific. High expression of endoglin in non-tumour tissue suggests that this microenvironment might play an especially important role in the progression of HCC. Evaluation of tissue expression, as well as serum concentrations of this glycoprotein in HCC, tends to confirm its role as an important biomarker in HCC diagnosis and prognosis. The role of endoglin in liver fibrosis and HCC progression also makes it an attractive therapeutic target. Despite these facts, the exact molecular mechanisms of endoglin functioning in hepatocarcinogenesis are still poorly understood. This review summarizes the current data concerning the role and signalling pathways of endoglin in hepatocellular carcinoma development and progression, and provides an overview of the strategies available for a specific targeting of CD105 in anti-angiogenic therapy in HCC.
Collapse
Affiliation(s)
- Aldona Kasprzak
- Department of Histology and Embryology, University of Medical Sciences, Poznań 60-781, Poland.
| | - Agnieszka Adamek
- Department of Infectious Diseases, Hepatology and Acquired Immunodeficiencies, University of Medical Sciences, Poznań 61-285, Poland.
| |
Collapse
|
28
|
Nickel AC, Wan XY, Saban DV, Weng YL, Zhang S, Keyvani K, Sure U, Zhu Y. Loss of programmed cell death 10 activates tumor cells and leads to temozolomide-resistance in glioblastoma. J Neurooncol 2018; 141:31-41. [PMID: 30392087 DOI: 10.1007/s11060-018-03017-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/25/2018] [Indexed: 11/24/2022]
Abstract
PURPOSE Glioblastoma (GBM) is one of the most aggressive and incurable primary brain tumors. Identification of novel therapeutic targets is an urgent priority. Programmed cell death 10 (PDCD10), a ubiquitously expressed apoptotic protein, has shown a dual function in different types of cancers and in chemo-resistance. Recently, we reported that PDCD10 was downregulated in human GBM. The aim of this study was to explore the function of PDCD10 in GBM cells. METHODS PDCD10 was knocked down in three GBM cell lines (U87, T98g and LN229) by lentiviral-mediated shRNA transduction. U87 and T98g transduced cells were used for phenotype study and LN229 and T98g cells were used for apoptosis study. The role of PDCD10 in apoptosis and chemo-resistance was investigated after treatment with staurosporine and temozolomide. A GBM xenograft mouse model was used to confirm the function of PDCD10 in vivo. A protein array was performed in PDCD10-knockdown and control GBM cells. RESULTS Knockdown of PDCD10 in GBM cells promoted cell proliferation, adhesion, migration, invasion, and inhibited apoptosis and caspase-3 activation. PDCD10-knockdown accelerated tumor growth and increased tumor mass by 2.1-fold and led to a chemo-resistance of mice treated with temozolomide. Immunostaining revealed extensive Ki67-positive cells and less activation of caspase-3 in PDCD10-knockdown tumors. The protein array demonstrated an increased release of multiple growth factors from PDCD10-knockdown GBM cells. CONCLUSIONS Loss of programmed cell death 10 activates tumor cells and leads to temozolomide-resistance in GBM, suggesting PDCD10 as a potential target for GBM therapy.
Collapse
Affiliation(s)
- Ann-Christin Nickel
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Xue-Yan Wan
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.,Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dino-Vitali Saban
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Yin-Lun Weng
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.,Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shu Zhang
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Ulrich Sure
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Yuan Zhu
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|
29
|
Tumor Targeting and Tumor Growth Inhibition Capability of Mesoporous Silica Nanoparticles in Mouse Models. Enzymes 2018; 44:61-82. [PMID: 30360815 DOI: 10.1016/bs.enz.2018.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mesoporous silica nanoparticles (MSNs) provide a promising vehicle for anticancer drug delivery. Various animal studies point to the efficacy of this nanoparticle for delivering anticancer agents (drug and siRNA) to inhibit tumor growth. These studies also showed tumor accumulation of MSN nanoparticles. While the extent of tumor accumulation differed, the study showed that it is possible to achieve significant accumulation of nanoparticles in the tumor. Biocompatibility and safety of MSN were also demonstrated by these studies.
Collapse
|
30
|
Choueiri TK, Michaelson MD, Posadas EM, Sonpavde GP, McDermott DF, Nixon AB, Liu Y, Yuan Z, Seon BK, Walsh M, Jivani MA, Adams BJ, Theuer CP. An Open Label Phase Ib Dose Escalation Study of TRC105 (Anti-Endoglin Antibody) with Axitinib in Patients with Metastatic Renal Cell Carcinoma. Oncologist 2018; 24:202-210. [PMID: 30190302 DOI: 10.1634/theoncologist.2018-0299] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/02/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND TRC105 is an IgG1 endoglin monoclonal antibody that potentiates VEGF inhibitors in preclinical models. We assessed safety, pharmacokinetics, and antitumor activity of TRC105 in combination with axitinib in patients with metastatic renal cell carcinoma (mRCC). SUBJECTS, MATERIALS, AND METHODS Heavily pretreated mRCC patients were treated with TRC105 weekly (8 mg/kg and then 10 mg/kg) in combination with axitinib (initially at 5 mg b.i.d. and then escalated per patient tolerance to a maximum of 10 mg b.i.d.) until disease progression or unacceptable toxicity using a standard 3 + 3 phase I design. RESULTS Eighteen patients (median number of prior therapies = 3) were treated. TRC105 dose escalation proceeded to 10 mg/kg weekly without dose-limiting toxicity. Adverse event characteristics of each drug were not increased in frequency or severity when the two drugs were administered concurrently. TRC105 and axitinib demonstrated preliminary evidence of activity, including partial responses (PR) by RECIST in 29% of patients, and median progression-free survival (11.3 months). None of the patients with PR had PR to prior first-line treatment. Lower baseline levels of osteopontin and higher baseline levels of TGF-β receptor 3 correlated with overall response rate. CONCLUSION TRC105 at 8 and 10 mg/kg weekly was well tolerated in combination with axitinib, with encouraging evidence of activity in patients with mRCC. A multicenter, randomized phase II trial of TRC105 and axitinib has recently completed enrollment (NCT01806064). IMPLICATIONS FOR PRACTICE TRC105 is a monoclonal antibody to endoglin (CD105), a receptor densely expressed on proliferating endothelial cells and also on renal cancer stem cells that is implicated as a mediator of resistance to inhibitors of the VEGF pathway. In this Phase I trial, TRC105 combined safely with axitinib at the recommended single agent doses of each drug in patients with renal cell carcinoma. The combination demonstrated durable activity in a VEGF inhibitor-refractory population and modulated several angiogenic biomarkers. A randomized Phase II trial testing TRC105 in combination with axitinib in clear cell renal cell carcinoma has completed accrual.
Collapse
Affiliation(s)
| | | | | | - Guru P Sonpavde
- University of Alabama Comprehensive Cancer Center, Birmingham, Alabama, USA
| | | | - Andrew B Nixon
- Duke University Medical Center, Durham, North Carolina, USA
| | - Yingmiao Liu
- Duke University Medical Center, Durham, North Carolina, USA
| | - Zhenhua Yuan
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ben K Seon
- Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Meghara Walsh
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Manoj A Jivani
- TRACON Pharmaceuticals, Inc., San Diego, California, USA
| | - Bonne J Adams
- TRACON Pharmaceuticals, Inc., San Diego, California, USA
| | | |
Collapse
|
31
|
Chen F, Goel S, Shi S, Barnhart TE, Lan X, Cai W. General synthesis of silica-based yolk/shell hybrid nanomaterials and in vivo tumor vasculature targeting. NANO RESEARCH 2018; 11:4890-4904. [PMID: 30410684 PMCID: PMC6217832 DOI: 10.1007/s12274-018-2078-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/14/2018] [Accepted: 04/17/2018] [Indexed: 05/23/2023]
Abstract
Multifunctional yolk/shell-structured hybrid nanomaterials have attracted increasing interest as theranostic nanoplatforms for cancer imaging and therapy. However, because of the lack of suitable surface engineering and tumor targeting strategies, previous research has focused mainly on nanostructure design and synthesis with few successful examples showing active tumor targeting after systemic administration. In this study, we report the general synthetic strategy of chelator-free zirconium-89 (89Zr)-radiolabeled, TRC105 antibody-conjugated, silica-based yolk/shell hybrid nanoparticles for in vivo tumor vasculature targeting. Three types of inorganic nanoparticles with varying morphologies and sizes were selected as the internal cores, which were encapsulated into single hollow mesoporous silica nanoshells to form the yolk/shell-structured hybrid nanoparticles. As a proof-of-concept, we demonstrated successful surface functionalization of the nanoparticles with polyethylene glycol, TRC105 antibody (specific forCD105/endoglin), and 89Zr (a positron-emitting radioisotope), and enhanced in vivo tumor vasculature-targeted positron emission tomography imaging in 4T1murine breast tumor-bearing mice. This strategy could be applied to the synthesis of other types of yolk/shell theranostic nanoparticles for tumor-targeted imaging and drug delivery.
Collapse
Affiliation(s)
- Feng Chen
- Department of Radiology, University of Wisconsin-Madison, WI 53705, USA
| | - Shreya Goel
- Materials Science Program, University of Wisconsin-Madison, WI 53705, USA
| | - Sixiang Shi
- Materials Science Program, University of Wisconsin-Madison, WI 53705, USA
| | - Todd E. Barnhart
- Department of Medical Physics, University of Wisconsin-Madison, WI 53705, USA
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weibo Cai
- Department of Radiology, University of Wisconsin-Madison, WI 53705, USA
- Materials Science Program, University of Wisconsin-Madison, WI 53705, USA
- Department of Medical Physics, University of Wisconsin-Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA
| |
Collapse
|
32
|
Hahn AW, Pal SK, Agarwal N. Targeting Endoglin to Treat Metastatic Renal Cell Carcinoma: Lessons from Osler-Weber-Rendu Syndrome. Oncologist 2018; 24:143-145. [PMID: 30139834 PMCID: PMC6369955 DOI: 10.1634/theoncologist.2018-0443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 01/12/2023] Open
Abstract
This commentary reviews the novel mechanism of action of TRC105 in the context of treatments currently available for metastatic renal cell carcinoma, highlighting the recent findings of Choueiri and colleagues.
Collapse
Affiliation(s)
- Andrew W Hahn
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Sumanta K Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, USA
| | - Neeraj Agarwal
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
33
|
Liu Y, Starr MD, Brady JC, Rushing C, Pang H, Adams B, Alvarez D, Theuer CP, Hurwitz HI, Nixon AB. Modulation of Circulating Protein Biomarkers in Cancer Patients Receiving Bevacizumab and the Anti-Endoglin Antibody, TRC105. Mol Cancer Ther 2018; 17:2248-2256. [DOI: 10.1158/1535-7163.mct-17-0916] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/23/2018] [Accepted: 07/06/2018] [Indexed: 11/16/2022]
|
34
|
de Bruin RG, Rabelink TJ, van Zonneveld AJ, van der Veer EP. Emerging roles for RNA-binding proteins as effectors and regulators of cardiovascular disease. Eur Heart J 2018; 38:1380-1388. [PMID: 28064149 DOI: 10.1093/eurheartj/ehw567] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 11/02/2016] [Indexed: 12/18/2022] Open
Abstract
The cardiovascular system comprises multiple cell types that possess the capacity to modulate their phenotype in response to acute or chronic injury. Transcriptional and post-transcriptional mechanisms play a key role in the regulation of remodelling and regenerative responses to damaged cardiovascular tissues. Simultaneously, insufficient regulation of cellular phenotype is tightly coupled with the persistence and exacerbation of cardiovascular disease. Recently, RNA-binding proteins such as Quaking, HuR, Muscleblind, and SRSF1 have emerged as pivotal regulators of these functional adaptations in the cardiovascular system by guiding a wide-ranging number of post-transcriptional events that dramatically impact RNA fate, including alternative splicing, stability, localization and translation. Moreover, homozygous disruption of RNA-binding protein genes is commonly associated with cardiac- and/or vascular complications. Here, we summarize the current knowledge on the versatile role of RNA-binding proteins in regulating the transcriptome during phenotype switching in cardiovascular health and disease. We also detail existing and potential DNA- and RNA-based therapeutic approaches that could impact the treatment of cardiovascular disease in the future.
Collapse
Affiliation(s)
- Ruben G de Bruin
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Ton J Rabelink
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Eric P van der Veer
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| |
Collapse
|
35
|
Wang X, Solban N, Khanna P, Callea M, Song J, Alsop DC, Pearsall RS, Atkins MB, Mier JW, Signoretti S, Alimzhanov M, Kumar R, Bhasin MK, Bhatt RS. Inhibition of ALK1 signaling with dalantercept combined with VEGFR TKI leads to tumor stasis in renal cell carcinoma. Oncotarget 2018; 7:41857-41869. [PMID: 27248821 PMCID: PMC5173101 DOI: 10.18632/oncotarget.9621] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/05/2016] [Indexed: 12/21/2022] Open
Abstract
Treatment of metastatic renal cell carcinoma (mRCC) with agents that block signaling through vascular endothelial growth factor receptor 2 (VEGFR2) induces disease regression or stabilization in some patients; however, these responses tend to be short-lived. Therefore, development of combination therapies that can extend the efficacy of VEGFR antagonists in mRCC remains a priority. We studied murine xenograft models of RCC that become refractory to treatment with the VEGFR tyrosine kinase inhibitor (TKI) sunitinib. Dalantercept is a novel antagonist of Activin receptor-like kinase 1 (ALK1)/Bone morphogenetic protein (BMP) 9 signaling. Dalantercept inhibited growth in the murine A498 xenograft model which correlated with hyperdilation of the tumor vasculature and an increase in tumor hypoxia. When combined with sunitinib, dalantercept induced tumor necrosis and prevented tumor regrowth and revascularization typically seen with sunitinib monotherapy in two RCC models. Combination therapy led to significant downregulation of angiogenic genes as well as downregulation of endothelial specific gene expression particularly of the Notch signaling pathway. We demonstrate that simultaneous targeting of molecules that control distinct phases of angiogenesis, such as ALK1 and VEGFR, is a valid strategy for treatment of mRCC. At the molecular level, combination therapy leads to downregulation of Notch signaling.
Collapse
Affiliation(s)
- Xiaoen Wang
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Prateek Khanna
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Marcella Callea
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jiaxi Song
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - David C Alsop
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Michael B Atkins
- Departments of Oncology and Medicine, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - James W Mier
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Ravi Kumar
- Acceleron Pharma, Inc., Cambridge, MA, USA
| | - Manoj K Bhasin
- Division of Interdisciplinary Medicine & Biotechnology, and Genomics, Proteomics, Bioinformatics and Systems Biology Center, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rupal S Bhatt
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
36
|
Di Paolo V, Russo I, Boldrini R, Ravà L, Pezzullo M, Benedetti MC, Galardi A, Colletti M, Rota R, Orlando D, Crocoli A, Peinado H, Milano GM, Di Giannatale A. Evaluation of Endoglin (CD105) expression in pediatric rhabdomyosarcoma. BMC Cancer 2018; 18:31. [PMID: 29304781 PMCID: PMC5755407 DOI: 10.1186/s12885-017-3947-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/20/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The Intratumoral Microvessel Density (IMVD) is commonly used to quantify tumoral vascularization and is usually assessed by pan-endothelial markers, such as CD31. Endoglin (CD105) is a protein predominantly expressed in proliferating endothelium and the IMVD determined by this marker measures specifically the neovascularization. In this study, we investigated the CD105 expression in pediatric rhabdomyosarcoma and assessed the neovascularization by using the angiogenic ratio IMVD-CD105 to IMVD-CD31. METHODS Paraffin-embedded archival tumor specimens were selected from 65 pediatric patients affected by rhabdomyosarcoma. The expression levels of CD105, CD31 and Vascular Endothelial Growth Factor (VEGF) were investigated in 30 cases (18 embryonal and 12 alveolar) available for this study. The IMVD-CD105 to IMVD-CD31 expression ratio was correlated with clinical and pathologic features of these patients. RESULTS We found a specific expression of endoglin (CD105) in endothelial cells of all the rhabdomyosarcoma specimens analyzed. We observed a significant positive correlation between the IMVD individually measured by CD105 and CD31. The CD105/CD31 expression ratio was significantly higher in patients with lower survival and embryonal histology. Indeed, patients with a CD105/CD31 expression ratio < 1.3 had a significantly increased OS (88%, 95%CI, 60%-97%) compared to patients with higher values (40%, 95%CI, 12%-67%). We did not find any statistical correlation among VEGF and EFS, OS and CD105/CD31 expression ratio. CONCLUSION CD105 is expressed on endothelial cells of rhabdomyosarcoma and represent a useful tool to quantify neovascularization in this tumor. If confirmed by further studies, these results will indicate that CD105 is a potential target for combined therapies in rhabdomyosarcoma.
Collapse
Affiliation(s)
- Virginia Di Paolo
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy
| | - Ida Russo
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy
| | - Renata Boldrini
- Department of Laboratories - Pathology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy
| | - Lucilla Ravà
- Clinical Epidemiology, Bambino Gesù Children’s Hospital, IRCCS, Viale Ferdinando Baldelli 41, 00146 Rome, Italy
| | - Marco Pezzullo
- Core Facilities, Bambino Gesù Children’s Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy
| | - Maria Chiara Benedetti
- Department of Laboratories - Pathology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy
| | - Angela Galardi
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy
| | - Marta Colletti
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy
| | - Rossella Rota
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy
| | - Domenico Orlando
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy
| | - Alessandro Crocoli
- General Pediatric and Thoracic Surgery, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy
| | - Hector Peinado
- Microenvironment and Metastasis Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), C/ Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Giuseppe Maria Milano
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy
| | - Angela Di Giannatale
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy
| |
Collapse
|
37
|
Shi S, Chen F, Goel S, Graves SA, Luo H, Theuer CP, Engle JW, Cai W. In Vivo Tumor-Targeted Dual-Modality PET/Optical Imaging with a Yolk/Shell-Structured Silica Nanosystem. NANO-MICRO LETTERS 2018; 10:65. [PMID: 30393713 PMCID: PMC6199109 DOI: 10.1007/s40820-018-0216-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/14/2018] [Indexed: 05/18/2023]
Abstract
Silica nanoparticles have been one of the most promising nanosystems for biomedical applications due to their facile surface chemistry and non-toxic nature. However, it is still challenging to effectively deliver them into tumor sites and noninvasively visualize their in vivo biodistribution with excellent sensitivity and accuracy for effective cancer diagnosis. In this study, we design a yolk/shell-structured silica nanosystem 64Cu-NOTA-QD@HMSN-PEG-TRC105, which can be employed for tumor vasculature targeting and dual-modality PET/optical imaging, leading to superior targeting specificity, excellent imaging capability and more reliable diagnostic outcomes. By combining vasculature targeting, pH-sensitive drug delivery, and dual-modality imaging into a single platform, as-designed yolk/shell-structured silica nanosystems may be employed for the future image-guided tumor-targeted drug delivery, to further enable cancer theranostics.
Collapse
Affiliation(s)
- Sixiang Shi
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Feng Chen
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, 53705-2275, USA
| | - Shreya Goel
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Stephen A Graves
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Haiming Luo
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, 53705-2275, USA
| | | | - Jonathan W Engle
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Weibo Cai
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, 53705-2275, USA.
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
| |
Collapse
|
38
|
Roman BL, Hinck AP. ALK1 signaling in development and disease: new paradigms. Cell Mol Life Sci 2017; 74:4539-4560. [PMID: 28871312 PMCID: PMC5687069 DOI: 10.1007/s00018-017-2636-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 08/01/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022]
Abstract
Activin A receptor like type 1 (ALK1) is a transmembrane serine/threonine receptor kinase in the transforming growth factor-beta receptor family that is expressed on endothelial cells. Defects in ALK1 signaling cause the autosomal dominant vascular disorder, hereditary hemorrhagic telangiectasia (HHT), which is characterized by development of direct connections between arteries and veins, or arteriovenous malformations (AVMs). Although previous studies have implicated ALK1 in various aspects of sprouting angiogenesis, including tip/stalk cell selection, migration, and proliferation, recent work suggests an intriguing role for ALK1 in transducing a flow-based signal that governs directed endothelial cell migration within patent, perfused vessels. In this review, we present an updated view of the mechanism of ALK1 signaling, put forth a unified hypothesis to explain the cellular missteps that lead to AVMs associated with ALK1 deficiency, and discuss emerging roles for ALK1 signaling in diseases beyond HHT.
Collapse
Affiliation(s)
- Beth L Roman
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto St, Pittsburgh, PA, 15261, USA.
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
39
|
Zhan Y, Shi S, Ehlerding EB, Graves SA, Goel S, Engle JW, Liang J, Tian J, Cai W. Radiolabeled, Antibody-Conjugated Manganese Oxide Nanoparticles for Tumor Vasculature Targeted Positron Emission Tomography and Magnetic Resonance Imaging. ACS APPLIED MATERIALS & INTERFACES 2017; 9:38304-38312. [PMID: 29028311 PMCID: PMC5680099 DOI: 10.1021/acsami.7b12216] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Manganese oxide nanoparticles (Mn3O4 NPs) have attracted a great deal of attention in the field of biomedical imaging because of their ability to create an enhanced imaging signal in MRI as novel potent T1 contrast agents. In this study, we present tumor vasculature-targeted imaging in mice using Mn3O4 NPs through conjugation to the anti-CD105 antibody TRC105 and radionuclide copper-64 (64Cu, t1/2: 12.7 h). The Mn3O4 conjugated NPs, 64Cu-NOTA-Mn3O4@PEG-TRC105, exhibited sufficient stability in vitro and in vivo. Serial positron emission tomography (PET) and magnetic resonance imaging (MRI) studies evaluated the pharmacokinetics and demonstrated targeting of 64Cu-NOTA-Mn3O4@PEG-TRC105 to 4T1 murine breast tumors in vivo, compared to 64Cu-NOTA-Mn3O4@PEG. The specificity of 64Cu-NOTA-Mn3O4@PEG-TRC105 for the vascular marker CD105 was confirmed through in vivo, in vitro, and ex vivo experiments. Since Mn3O4 conjugated NPs exhibited desirable properties for T1 enhanced imaging and low toxicity, the tumor-specific Mn3O4 conjugated NPs reported in this study may serve as promising multifunctional nanoplatforms for precise cancer imaging and diagnosis.
Collapse
Affiliation(s)
- Yonghua Zhan
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, 710071, China
| | - Sixiang Shi
- Department of Radiology, University of Wisconsin–Madison, WI, 53705,USA
| | - Emily B. Ehlerding
- Department of Medical Physics, University of Wisconsin-Madison, WI, 53705,USA
| | - Stephen A. Graves
- Department of Medical Physics, University of Wisconsin-Madison, WI, 53705,USA
| | - Shreya Goel
- Department of Radiology, University of Wisconsin–Madison, WI, 53705,USA
| | - Jonathan W. Engle
- Department of Medical Physics, University of Wisconsin-Madison, WI, 53705,USA
| | - Jimin Liang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, 710071, China
- Department of Medical Physics, University of Wisconsin-Madison, WI, 53705,USA
| | - Jie Tian
- Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- Corresponding Authors: (W. Cai); (J. Tian)
| | - Weibo Cai
- Department of Radiology, University of Wisconsin–Madison, WI, 53705,USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, 53705, USA
- Corresponding Authors: (W. Cai); (J. Tian)
| |
Collapse
|
40
|
Tan J, Yang N, Zhong L, Tan J, Hu Z, Zhao Q, Gong W, Zhang Z, Zheng R, Lai Z, Li Y, Zhou C, Zhang G, Zheng D, Zhang Y, Wu S, Jiang X, Zhong J, Huang Y, Zhou S, Zhao Y. A New Theranostic System Based on Endoglin Aptamer Conjugated Fluorescent Silica Nanoparticles. Theranostics 2017; 7:4862-4876. [PMID: 29187909 PMCID: PMC5706105 DOI: 10.7150/thno.19101] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 08/11/2017] [Indexed: 12/14/2022] Open
Abstract
Background: Tumor vessels can potentially serve as diagnostic, prognostic and therapeutic targets for solid tumors. Fluorescent dyes are commonly used as biological indicators, while photobleaching seriously hinders their application. In this study, we aim to generate a fluorescent silica nanoparticles (FSiNPs) theranostic system marked by the mouse endgolin (mEND) aptamer, YQ26. Methods: A highly specific YQ26 was selected by using gene-modified cell line-based SELEX technique. FSiNPs were prepared via the reverse microemulsion method. The YQ26-FSiNPs theranostic system was developed by combining YQ26 with the FSiNPs for in vivo tumor imaging, treatment and monitoring. Results: Both in vitro experiments (i.e. cellular and tumor tissue targeting assays) and in vivo animal studies (i.e. in vivo imaging and antitumor efficacy of YQ26-FSiNPs) clearly demonstrated that YQ26-FSiNPs could achieve prominently high targeting efficiency and therapeutic effects via aptamer YQ26-mediated binding to endoglin (END) molecule. Conclusion: This simple, sensitive, and specific YQ26-FSiNPs theranostic system has a great potential for clinical tumor targeting imaging and treatment.
Collapse
Affiliation(s)
- Juntao Tan
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Nuo Yang
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Liping Zhong
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jie Tan
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zixi Hu
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Qing Zhao
- Department of Theracic Surgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, China
| | - Wenlin Gong
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhenghua Zhang
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Rong Zheng
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zongqiang Lai
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yanmei Li
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Chaofan Zhou
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Guoqing Zhang
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Duo Zheng
- Shenzhen Key Laboratory of Translational Medicine of Tumor, Department of Basic Medicine, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518000, China
| | - Ying Zhang
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Siyu Wu
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xinglu Jiang
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jianhong Zhong
- Department of Surgery Oncology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yong Huang
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Sufang Zhou
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yongxiang Zhao
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| |
Collapse
|
41
|
Dorff TB, Longmate JA, Pal SK, Stadler WM, Fishman MN, Vaishampayan UN, Rao A, Pinksi JK, Hu JS, Quinn DI, Lara PN. Bevacizumab alone or in combination with TRC105 for patients with refractory metastatic renal cell cancer. Cancer 2017; 123:4566-4573. [PMID: 28832978 DOI: 10.1002/cncr.30942] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/19/2017] [Accepted: 07/11/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND Targeting the vascular endothelial growth factor (VEGF) pathway has improved outcomes in metastatic renal cell carcinoma (RCC); however, resistance inevitably occurs. CD105 (endoglin) is an angiogenic pathway that is strongly upregulated after VEGF inhibition, potentially contributing to resistance. The authors tested whether TRC105, a monoclonal antibody against endoglin, impacted disease control in patients with previously treated RCC who were receiving bevacizumab. METHODS Eligible patients with metastatic RCC who had previously received 1 to 4 prior lines of therapy, including VEGF-targeted agents, were randomized 1:1 to receive bevacizumab 10 mg/kg intravenously every 2 weeks (arm A) or the same plus TRC105 10 mg/kg intravenously every 2 weeks (arm B). The primary endpoint was progression-free survival (PFS) at 12 and 24 weeks. Correlative studies included serum transforming growth factor β (TGFβ) and CD105 levels as well as tissue immunostaining for TGFβ receptors. RESULTS Fifty-nine patients were enrolled (28 on arm A and 31 on arm B), and 1 patient on each arm had a confirmed partial response. The median PFS for bevacizumab alone was 4.6 months compared with 2.8 for bevacizumab plus TRC105 (P = .09). Grade ≥ 3 toxicities occurred in 16 patients (57%) who received bevacizumab compared with 19 (61%) who received bevacizumab plus TRC105 (P = .9). Baseline serum TGFβ levels below the median (<10.6 ng/mL) were associated with longer median PFS (5.6 vs 2.1 months; P = .014). CONCLUSIONS TRC105 failed to improve PFS when added to bevacizumab. TGFβ warrants further study as a biomarker in RCC. Cancer 2017;123:4566-4573. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Tanya B Dorff
- University of Southern California Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, California
| | - Jeff A Longmate
- Department of Biostatistics, City of Hope, Duarte, California
| | - Sumanta K Pal
- Department of Medical Oncology and Experimental Therapeutics, City of Hope, Duarte, California
| | - Walter M Stadler
- Hematology/Oncology Section, University of Chicago, Chicago, Illinois
| | - Mayer N Fishman
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Ulka N Vaishampayan
- Department of Hematology and Oncology, Karmanos Cancer Center, Detroit, Michigan
| | - Amol Rao
- University of Southern California Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, California
| | - Jacek K Pinksi
- University of Southern California Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, California
| | - James S Hu
- University of Southern California Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, California
| | - David I Quinn
- Department of Medical Oncology, University of Southern California Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, California
| | - Primo N Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, California
| |
Collapse
|
42
|
Posadas EM, Limvorasak S, Figlin RA. Targeted therapies for renal cell carcinoma. Nat Rev Nephrol 2017; 13:496-511. [DOI: 10.1038/nrneph.2017.82] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
43
|
Abstract
OBJECTIVE The objective of this study was to determine the contribution of lymphatic tissue to heterotopic ossification (HO). BACKGROUND HO is the pathologic development of ectopic bone within soft tissues often following severe trauma. Characterization of the tissue niche supporting HO is critical to identifying therapies directed against this condition. Lymphangiogenesis is upregulated during incidents of trauma, thereby coincident with the niche supportive of HO. We hypothesized that lymphatic tissues play a critical role in HO formation. METHODS Mice underwent hindlimb Achilles' tendon transection and dorsal burn injury (burn/tenotomy) to induce HO. The popliteal and inguinal lymph nodes were excised ipsilateral to the tenotomy site. Flow cytometry and immunostaining were used to quantify and localize lymphoendothelium. MicroCT was used to quantify HO. RESULTS Enrichment of mature lymphatic tissues was noted 2 weeks after injury at the tendon transection sites when compared with the contralateral, intact tendon based on LYVE1+ tubules (10.9% vs 0.8%, P < 0.05). Excision of the inguinal and popliteal nodes with draining popliteal lymphatic vessel significantly decreased the presence of mature lymphoendothelium 2 weeks after injury (10.9% vs 3.3%, P < 0.05). Bone-cartilage-stromal progenitor cells (CD105+/AlphaV+/Tie2-/CD45-/CD90-/BP1-) were also significantly decreased after lymph node excision (10.2% vs 0.5%, P < 0.05). A significant decrease was noted in the volume of de novo HO present within the soft tissues (0.12 mm vs 0.02 mm). CONCLUSION These findings suggest that lymphatic vessels are intimately linked with the de novo formation bone within soft tissues following trauma, and their presence may facilitate bone formation.
Collapse
|
44
|
Rakocevic J, Orlic D, Mitrovic-Ajtic O, Tomasevic M, Dobric M, Zlatic N, Milasinovic D, Stankovic G, Ostojić M, Labudovic-Borovic M. Endothelial cell markers from clinician's perspective. Exp Mol Pathol 2017; 102:303-313. [PMID: 28192087 DOI: 10.1016/j.yexmp.2017.02.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/04/2017] [Accepted: 02/08/2017] [Indexed: 02/07/2023]
Abstract
Endothelial cell markers are membrane-bound or cytoplasmic molecules expressed by endothelial cells, which help their easier identification and discrimination from other cell types. During vasculogenesis, endothelial cells differentiate from hemangioblasts to form new blood vessels. With the discovery of endothelial progenitor cells (EPC) and their ability to form new blood vessels, the term vasculogenesis is not only reserved for the embryonic development. Possibility of de novo blood vessel formation from EPC is now widely explored in different ischemic conditions, especially in cardiovascular medicine. Numerous clinical trials have tested enhancing tissue vascularization by delivering hematopoietic cells that expressed endothelial markers. This therapeutic approach proved to be challenging and promising, particularly for patients who have exhausted all conventional therapeutic modalities. Angiogenesis, which refers to the formation of new blood vessels from existing vasculature, is indispensable process during tumor progression and metastasis. Blockage of tumor angiogenesis by targeting and inhibiting endothelial cell has emerged as novel safe and efficacious method to control many advanced malignant diseases. Numerous clinical studies are currently testing new antiangiogenic drugs which target and inhibit endothelial cell markers, receptors or molecules which transmit receptor-mediated signals, therefore inhibiting endothelial cell proliferation, migration and vascular tube formation. Many of these drugs are now widely used in clinical settings as first- or second-line chemotherapy in advanced malignant conditions. So far, these therapeutic approaches gave modest, yet encouraging clinical improvements, prolonging survival and improving functional capacity and quality of life for many terminally ill patients. Here we present the most commonly used endothelial cell markers along with their applicability in contemporary clinical practice.
Collapse
Affiliation(s)
- Jelena Rakocevic
- Institute of Histology and Embryology, School of Medicine, University of Belgrade, 26 Visegradska Street, Belgrade, Serbia
| | - Dejan Orlic
- Cardiology Clinic, Clinical Center of Serbia, 8 Dr Koste Todorovica Street, Belgrade, Serbia; School of Medicine, University of Belgrade, 8 Dr Subotica Street, Belgrade, Serbia
| | - Olivera Mitrovic-Ajtic
- Department for Neuroendocrinology, Institute for Medical Research, 4 Dr Subotica Street, Belgrade, Serbia
| | - Miloje Tomasevic
- Cardiology Clinic, Clinical Center of Serbia, 8 Dr Koste Todorovica Street, Belgrade, Serbia; Department of Internal medicine, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, Kragujevac, Serbia
| | - Milan Dobric
- Cardiology Clinic, Clinical Center of Serbia, 8 Dr Koste Todorovica Street, Belgrade, Serbia; School of Medicine, University of Belgrade, 8 Dr Subotica Street, Belgrade, Serbia
| | - Natasa Zlatic
- School of Medicine, University of Belgrade, 8 Dr Subotica Street, Belgrade, Serbia
| | - Dejan Milasinovic
- Cardiology Clinic, Clinical Center of Serbia, 8 Dr Koste Todorovica Street, Belgrade, Serbia
| | - Goran Stankovic
- Cardiology Clinic, Clinical Center of Serbia, 8 Dr Koste Todorovica Street, Belgrade, Serbia; School of Medicine, University of Belgrade, 8 Dr Subotica Street, Belgrade, Serbia
| | - Miodrag Ostojić
- School of Medicine, University of Belgrade, 8 Dr Subotica Street, Belgrade, Serbia
| | - Milica Labudovic-Borovic
- Institute of Histology and Embryology, School of Medicine, University of Belgrade, 26 Visegradska Street, Belgrade, Serbia.
| |
Collapse
|
45
|
Núñez-Gómez E, Pericacho M, Ollauri-Ibáñez C, Bernabéu C, López-Novoa JM. The role of endoglin in post-ischemic revascularization. Angiogenesis 2016; 20:1-24. [PMID: 27943030 DOI: 10.1007/s10456-016-9535-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Following arterial occlusion, blood vessels respond by forming a new network of functional capillaries (angiogenesis), by reorganizing preexisting capillaries through the recruitment of smooth muscle cells to generate new arteries (arteriogenesis) and by growing and remodeling preexisting collateral arterioles into physiologically relevant arteries (collateral development). All these processes result in the recovery of organ perfusion. The importance of endoglin in post-occlusion reperfusion is sustained by several observations: (1) endoglin expression is increased in vessels showing active angiogenesis/remodeling; (2) genetic endoglin haploinsufficiency in humans causes deficient angiogenesis; and (3) the reduction of endoglin expression by gene disruption or the administration of endoglin-neutralizing antibodies reduces angiogenesis and revascularization. However, the precise role of endoglin in the several processes associated with revascularization has not been completely elucidated and, in some cases, the function ascribed to endoglin by different authors is controversial. The purpose of this review is to organize in a critical way the information available for the role of endoglin in several phenomena (angiogenesis, arteriogenesis and collateral development) associated with post-ischemic revascularization.
Collapse
Affiliation(s)
- Elena Núñez-Gómez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Claudia Ollauri-Ibáñez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Carmelo Bernabéu
- Centro de Investigaciones Biológicas, Spanish National Research Council (CIB, CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - José M López-Novoa
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain. .,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
46
|
Yuan ZX, Mo J, Zhao G, Shu G, Fu HL, Zhao W. Targeting Strategies for Renal Cell Carcinoma: From Renal Cancer Cells to Renal Cancer Stem Cells. Front Pharmacol 2016; 7:423. [PMID: 27891093 PMCID: PMC5103413 DOI: 10.3389/fphar.2016.00423] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/25/2016] [Indexed: 12/14/2022] Open
Abstract
Renal cell carcinoma (RCC) is a common form of urologic tumor that originates from the highly heterogeneous epithelium of renal tubules. Over the last decade, targeting therapies to renal cancer cells have transformed clinical care for RCC. Recently, it was proposed that renal cancer stem cells (CSCs) isolated from renal carcinomas were responsible for driving tumor growth and resistance to conventional chemotherapy and radiotherapy, according to the theory of CSCs; this has provided the rationale for therapies targeting this aggressive cell population. Precise identification of renal CSC populations and the complete cell hierarchy will accurately inform characterization of disease subtypes. This will ultimately contribute to more personalized and targeted therapies. Here, we summarize potential targeting strategies for renal cancer cells and renal CSCs, including tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors (mTOR), interleukins, CSC marker inhibitors, bone morphogenetic protein-2, antibody drug conjugates, and nanomedicine. In conclusion, targeting therapies for RCC represent new directions for exploration and clinical investigation and they plant a seed of hope for advanced clinical care.
Collapse
Affiliation(s)
- Zhi-Xiang Yuan
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Jingxin Mo
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Guixian Zhao
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Gang Shu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Hua-Lin Fu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Wei Zhao
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|
47
|
Orecchioni M, Ménard-Moyon C, Delogu LG, Bianco A. Graphene and the immune system: Challenges and potentiality. Adv Drug Deliv Rev 2016; 105:163-175. [PMID: 27235665 DOI: 10.1016/j.addr.2016.05.014] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/07/2016] [Accepted: 05/17/2016] [Indexed: 11/19/2022]
Abstract
In the growing area of nanomedicine, graphene-based materials (GBMs) are some of the most recent explored nanomaterials. For the majority of GBM applications in nanomedicine, the immune system plays a fundamental role. It is necessary to well understand the complexity of the interactions between GBMs, the immune cells, and the immune components and how they could be of advantage for novel effective diagnostic and therapeutic approaches. In this review, we aimed at painting the current picture of GBMs in the background of the immune system. The picture we have drawn looks like a cubist image, a sort of Picasso-like portrait looking at the topic from all perspectives: the challenges (due to the potential toxicity) and the potentiality like the conjugation of GBMs to biomolecules to develop advanced nanomedicine tools. In this context, we have described and discussed i) the impact of graphene on immune cells, ii) graphene as immunobiosensor, and iii) antibodies conjugated to graphene for tumor targeting. Thanks to the huge advances on graphene research, it seems realistic to hypothesize in the near future that some graphene immunoconjugates, endowed of defined immune properties, can go through preclinical test and be successfully used in nanomedicine.
Collapse
Affiliation(s)
- Marco Orecchioni
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Cécilia Ménard-Moyon
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Laboratoire d'Immunopathologie et de Chimie Thérapeutique, 67000 Strasbourg, France
| | - Lucia Gemma Delogu
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy.
| | - Alberto Bianco
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Laboratoire d'Immunopathologie et de Chimie Thérapeutique, 67000 Strasbourg, France.
| |
Collapse
|
48
|
Ma Y, Huang J, Song S, Chen H, Zhang Z. Cancer-Targeted Nanotheranostics: Recent Advances and Perspectives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:4936-4954. [PMID: 27150247 DOI: 10.1002/smll.201600635] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/22/2016] [Indexed: 05/10/2023]
Abstract
Cancer-targeted nanotechnology is experiencing the trend of finding new materials with multiple functions for imaging and therapeutic applications. With the rapid development of the related fields, there exists a large number of reports regarding theranostic nanomedicine, decreasing the gap between cancer diagnosis and treatment with minimized separate comprehensions. In order to present an overview on the cancer-targeted nanotheranostics, we first describe their essential building blocks, including platforms, therapeutic agents and imaging agents, and then the recently rapidly developed multimodal theranostic systems. Finally we discuss the major challenges and the perspectives of future development of nanotheranostics toward clinical translations and personalized nanomedicine.
Collapse
Affiliation(s)
- Yufei Ma
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, CAS Center for Excellence in Nanoscience, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jie Huang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, CAS Center for Excellence in Nanoscience, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Saijie Song
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, CAS Center for Excellence in Nanoscience, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Huabing Chen
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Zhijun Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, CAS Center for Excellence in Nanoscience, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| |
Collapse
|
49
|
Barriuso B, Antolín P, Arias FJ, Girotti A, Jiménez P, Cordoba-Diaz M, Cordoba-Diaz D, Girbés T. Anti-Human Endoglin (hCD105) Immunotoxin-Containing Recombinant Single Chain Ribosome-Inactivating Protein Musarmin 1. Toxins (Basel) 2016; 8:E184. [PMID: 27294959 PMCID: PMC4926150 DOI: 10.3390/toxins8060184] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/24/2016] [Accepted: 06/03/2016] [Indexed: 12/25/2022] Open
Abstract
Endoglin (CD105) is an accessory component of the TGF-β receptor complex, which is expressed in a number of tissues and over-expressed in the endothelial cells of tumor neovasculature. Targeting endoglin with immunotoxins containing type 2 ribosome-inactivating proteins has proved an effective tool to reduce blood supply to B16 mice tumor xenografts. We prepared anti-endoglin immunotoxin (IT)-containing recombinant musarmin 1 (single chain ribosome-inactivating proteins) linked to the mouse anti-human CD105 44G4 mouse monoclonal antibody via N-succinimidyl 3-(2-pyridyldithio) propionate (SPDP). The immunotoxin specifically killed L929 fibroblast mouse cells transfected with the short form of human endoglin with IC50 values in the range of 5 × 10(-10) to 10(-9) M.
Collapse
Affiliation(s)
- Begoña Barriuso
- Department of Biochemistry and Molecular Biology, University of Valladolid, 47005 Valladolid, Spain.
| | - Pilar Antolín
- Department of Biochemistry and Molecular Biology, University of Valladolid, 47005 Valladolid, Spain.
| | - F Javier Arias
- Department of Biochemistry and Molecular Biology, University of Valladolid, 47005 Valladolid, Spain.
- Bioforge, University of Valladolid, Spain and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 47002 Valladolid, Spain.
| | - Alessandra Girotti
- Bioforge, University of Valladolid, Spain and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 47002 Valladolid, Spain.
| | - Pilar Jiménez
- Department of Nutrition and Bromatology, University of Valladolid, 47005 Valladolid, Spain.
| | - Manuel Cordoba-Diaz
- Department of Pharmacy & Pharmaceutical Technology and University Institute of Industrial Pharmacy (IUFI), Complutense University of Madrid, 28040 Madrid, Spain.
| | - Damián Cordoba-Diaz
- Department of Pharmacy & Pharmaceutical Technology and University Institute of Industrial Pharmacy (IUFI), Complutense University of Madrid, 28040 Madrid, Spain.
| | - Tomás Girbés
- Department of Nutrition and Bromatology, University of Valladolid, 47005 Valladolid, Spain.
| |
Collapse
|
50
|
Modena A, Massari F, Ciccarese C, Brunelli M, Santoni M, Montironi R, Martignoni G, Tortora G. Targeting Met and VEGFR Axis in Metastatic Castration-Resistant Prostate Cancer: ‘Game Over’? Target Oncol 2016; 11:431-46. [DOI: 10.1007/s11523-015-0412-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|