1
|
Sağıroğlu S, Şirin C, Turgut AÇ, Tomruk C, Tuzcu A, Ertekin E, Uyanıkgil Y, Turgut M. Investigation of the Efficacy of Bevacizumab Treatment in An Experimental Rat Model of Chronic Subdural Hematoma. World Neurosurg 2024; 189:e272-e286. [PMID: 38871290 DOI: 10.1016/j.wneu.2024.06.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024]
Abstract
INTRODUCTION Chronic subdural hematoma (cSDH), a condition that develops over time, is characterized by inflammation, angiogenesis, and membrane development. As the population's average age increases, the incidence of cSDH is expected to grow. While surgery is the primary treatment technique, medicinal therapy options are being explored for high-risk patients. Currently, the most effective therapy combination is dexamethasone (Dex) and atorvastatin (Ato); however, it is associated with an increased risk of mortality. This study explored the effects of bevacizumab (Bev), a vascular endothelial growth factor antagonist, on cSDH. MATERIALS AND METHODS Ninety-five rats were divided into four groups (n = 18): sham, control hematoma, Dex-Ato, and Bev. Two separate autologous blood injections into the subdural space were used as the model. Weight was monitored for all rats to assess changes in their overall health. The control group was given i.p. saline, the Dex-Ato treatment was given by gavage, and the Bev treatment was given i.p. On seventh, 14th and 21st days six rats from each group were sacrificed and analyzed, while 23 rats were excluded from the experiment. RESULTS The maximum immunological response to cSDH was observed on day 14. Hematoma volume decreased over time in all groups. Dex-Ato and Bev were both found effective, while Dex-Ato caused weight loss. CONCLUSION Bev had similar effects to the Dex-Ato group and was well tolerated by rats. Given that cSDH is a disease of the elderly and vulnerable populations, Bev may be a viable alternative that can shed light on the disease's etiology for future research.
Collapse
Affiliation(s)
- Sinan Sağıroğlu
- Department of Neurosurgery, Aydın Adnan Menderes University Faculty of Medicine, Aydın, Turkey
| | - Cansın Şirin
- Department of Histology and Embryology, Ege University Faculty of Medicine, İzmir, Turkey
| | - Ali Çağlar Turgut
- Department of Radiology, Ege University Faculty of Medicine, İzmir, Turkey; Department of Histology and Embryology, Aydın Adnan Menderes University Health Sciences Institute, Aydın, Turkey
| | - Canberk Tomruk
- Department of Histology and Embryology, Samsun Education and Research Hospital, Samsun, Turkey
| | - Ayça Tuzcu
- Department of Biochemistry, Aydın Adnan Menderes University Faculty of Medicine, Aydın, Turkey
| | - Ersen Ertekin
- Department of Radiology, Hitit University Faculty of Medicine, Çorum, Turkey
| | - Yiğit Uyanıkgil
- Department of Histology and Embryology, Ege University Faculty of Medicine, İzmir, Turkey; Department of Stem Cell, Ege University, Health Science Institute, İzmir, Turkey; Cord Blood, Cell and Tissue Research and Application Centre, Ege University, İzmir, Turkey
| | - Mehmet Turgut
- Department of Neurosurgery, Aydın Adnan Menderes University Faculty of Medicine, Aydın, Turkey; Department of Histology and Embryology, Aydın Adnan Menderes University Health Sciences Institute, Aydın, Turkey.
| |
Collapse
|
2
|
Kumar A, Nagasaka Y, Jayananthan V, Zidan A, Heisler-Taylor T, Ambati J, Tamiya S, Kerur N. Therapeutic targeting of telomerase ameliorates experimental choroidal neovascularization. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167156. [PMID: 38582267 PMCID: PMC11497592 DOI: 10.1016/j.bbadis.2024.167156] [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: 09/19/2023] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/08/2024]
Abstract
Choroidal neovascularization (CNV) is the principal driver of blindness in neovascular age-related macular degeneration (nvAMD). Increased activity of telomerase, has been associated with endothelial cell proliferation, survival, migration, and invasion in the context of tumor angiogenesis. Expanding on this knowledge, we investigated the role of telomerase in the development of CNV in mouse model. We observed increased gene expression and activity of telomerase in mouse CNV. Genetic deficiency of the telomerase components, telomerase reverse transcriptase (Tert) and telomerase RNA component (Terc) suppressed laser-induced CNV in mice. Similarly, a small molecule inhibitor of TERT (BIBR 1532), and antisense oligonucleotides (ASOs) targeting Tert and Terc reduced CNV growth. Bone marrow chimera studies suggested that telomerase activity in non-bone marrow-derived cells is crucial for the development of CNV. Comparison of BIBR 1532 with VEGF neutralizing therapeutic strategy in mouse revealed a comparable level of angiosuppressive activity. However, when BIBR and anti-VEGF antibodies were administered as a combination at sub-therapeutic doses, a statistically significant suppression of CNV was observed. These findings underscore the potential benefits of combining sub-therapeutic doses of BIBR and anti-VEGF antibodies for developing newer therapeutic strategies for NV-AMD. Telomerase inhibition with BIBR 1532 suppressed induction of multiple cytokines and growth factors critical for neovascularization. In conclusion, our study identifies telomerase as a promising therapeutic target for treating neovascular disease of the eye and thus provides a proof of principle for further exploration of telomerase inhibition as a novel treatment strategy for nvAMD.
Collapse
Affiliation(s)
- Aman Kumar
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yosuke Nagasaka
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Vinodhini Jayananthan
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Asmaa Zidan
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Tyler Heisler-Taylor
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jayakrishna Ambati
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Shigeo Tamiya
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Nagaraj Kerur
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
3
|
Yee EJ, Vigil I, Sun Y, Torphy RJ, Schulick RD, Zhu Y. Group XIV C-type lectins: emerging targets in tumor angiogenesis. Angiogenesis 2024; 27:173-192. [PMID: 38468017 PMCID: PMC11021320 DOI: 10.1007/s10456-024-09907-x] [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: 10/25/2023] [Accepted: 01/23/2024] [Indexed: 03/13/2024]
Abstract
C-type lectins, distinguished by a C-type lectin binding domain (CTLD), are an evolutionarily conserved superfamily of glycoproteins that are implicated in a broad range of physiologic processes. The group XIV subfamily of CTLDs are comprised of CD93, CD248/endosialin, CLEC14a, and thrombomodulin/CD141, and have important roles in creating and maintaining blood vessels, organizing extracellular matrix, and balancing pro- and anti-coagulative processes. As such, dysregulation in the expression and downstream signaling pathways of these proteins often lead to clinically relevant pathology. Recently, group XIV CTLDs have been shown to play significant roles in cancer progression, namely tumor angiogenesis and metastatic dissemination. Interest in therapeutically targeting tumor vasculature is increasing and the search for novel angiogenic targets is ongoing. Group XIV CTLDs have emerged as key moderators of tumor angiogenesis and metastasis, thus offering substantial therapeutic promise for the clinic. Herein, we review our current knowledge of group XIV CTLDs, discuss each's role in malignancy and associated potential therapeutic avenues, briefly discuss group XIV CTLDs in the context of two other relevant lectin families, and offer future direction in further elucidating mechanisms by which these proteins function and facilitate tumor growth.
Collapse
Affiliation(s)
- Elliott J Yee
- Department of Surgery, University of Colorado Anschutz Medical Campus, 12800 E 19th Avenue, RC1-North, P18-8116, Aurora, CO, 80045, USA
| | - Isaac Vigil
- Department of Surgery, University of Colorado Anschutz Medical Campus, 12800 E 19th Avenue, RC1-North, P18-8116, Aurora, CO, 80045, USA
| | - Yi Sun
- Department of Surgery, University of Colorado Anschutz Medical Campus, 12800 E 19th Avenue, RC1-North, P18-8116, Aurora, CO, 80045, USA
| | - Robert J Torphy
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Richard D Schulick
- Department of Surgery, University of Colorado Anschutz Medical Campus, 12800 E 19th Avenue, RC1-North, P18-8116, Aurora, CO, 80045, USA
| | - Yuwen Zhu
- Department of Surgery, University of Colorado Anschutz Medical Campus, 12800 E 19th Avenue, RC1-North, P18-8116, Aurora, CO, 80045, USA.
| |
Collapse
|
4
|
Parsons NB, Annamalai B, Rohrer B. Regulatable Complement Inhibition of the Alternative Pathway Mitigates Wet Age-Related Macular Degeneration Pathology in a Mouse Model. Transl Vis Sci Technol 2023; 12:17. [PMID: 37462980 PMCID: PMC10362922 DOI: 10.1167/tvst.12.7.17] [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: 03/21/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
Purpose Risk for developing age-related macular degeneration (AMD) is linked to an overactive complement system. In the mouse model of laser-induced choroidal neovascularization (CNV), elevated levels of complement effector molecules, including complement C3, have been identified, and the alternative pathway (AP) is required for pathology. The main soluble AP regular is complement factor H (fH). We have previously shown that AP inhibition via subretinal AAV-mediated delivery of CR2-fH using a constitutive promoter is efficacious in reducing CNV. Here we ask whether the C3 promoter (pC3) effectively drives CR2-fH bioavailability for gene therapy. Methods Truncated pC3 was used to generate plasmids pC3-mCherry/CR2-fH followed by production of corresponding AAV5 vectors. pC3 activation was determined in transiently transfected ARPE-19 cells stimulated with H2O2 or normal human serum (+/- antioxidant or humanized CR2-fH, respectively). CNV was analyzed in C57BL/6J mice treated subretinally with AAV5-pC3-mCherry/CR2-fH using imaging (optical coherence tomography [OCT] and fundus imaging), functional (electroretinography [ERG]), and molecular (protein expression) readouts. Results Modulation of pC3 in vitro is complement and oxidative stress dependent, as shown by mCherry fluorescence. AAV5-pC3-CR2-fH were identified as safe and effective using OCT and ERG. CR2-fH expression significantly reduced CNV compared to mCherry and was correlated with reduced levels of C3dg/C3d in the retinal pigment epithelium/choroid fraction. Conclusions We conclude that complement-dependent regulation of AP inhibition ameliorates AMD pathology as effectively as using a constitutive promoter. Translational Relevance The goal of anticomplement therapy is to restore homeostatic levels of complement activation, which might be more easily achievable using a self-regulating system.
Collapse
Affiliation(s)
- Nathaniel B. Parsons
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA
| | | | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Division of Research, Charleston, SC, USA
| |
Collapse
|
5
|
Ruiz M, Zhang N, Sood AK, An Z. Antibody therapeutics for epithelial ovarian cancer. Expert Opin Biol Ther 2022; 22:1379-1391. [PMID: 36302510 PMCID: PMC10375545 DOI: 10.1080/14712598.2022.2141565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION High-grade serous ovarian carcinoma (HGSC) is an aggressive subtype of epithelial ovarian carcinoma (EOC) and remains the most lethal gynecologic cancer. A lack of effective and tolerable therapeutic options and nonspecific symptoms at presentation with advanced stage of disease are among the challenges in the management of the disease. AREAS COVERED An overview of ovarian cancer, followed by a discussion of the current therapeutic regimes and challenges that arise during and after the treatment of EOC. We discuss different formats of antibody therapeutics and their usage in targeting validated targets implicated in ovarian cancer, as well as three emerging novel proteins as examples recently implicated in their contribution to adaptive resistance in ovarian cancer. EXPERT OPINION Antibody therapeutics allow for a unique and effective way to target proteins implicated in cancer and other diseases, and have the potential to radically change the outcomes of patients suffering from ovarian cancer. The vast array of targets that have been implicated in ovarian cancer and yet the lack of effective therapeutic options for patients further stresses the importance of discovering novel proteins that can be targeted, as well as predictive biomarkers that can inform the stratification of patients into treatment-specific populations.
Collapse
Affiliation(s)
- Mason Ruiz
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
6
|
Attia SA, MacKay JA. Protein and polypeptide mediated delivery to the eye. Adv Drug Deliv Rev 2022; 188:114441. [PMID: 35817213 PMCID: PMC10049092 DOI: 10.1016/j.addr.2022.114441] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/23/2022]
Abstract
Hybrid or recombinant protein-polymers, peptide-based biomaterials, and antibody-targeted therapeutics are widely explored for various ocular conditions and vision correction. They have been noted for their potential biocompatibility, potency, adaptability, and opportunities for sustained drug delivery. Unique to peptide and protein therapeutics, their production by cellular translation allows their precise modification through genetic engineering. To a greater extent than drug delivery to other systems, delivery to the eye can benefit from the combination of locally-targeted administration and protein-based specificity. Consequently, a range of delivery platforms and administration methods have been exploited to address the ocular delivery of peptide and protein biomaterials. This review discusses a sample of preclinical and clinical opportunities for peptide-based drug delivery to the eye.
Collapse
Affiliation(s)
- Sara Aly Attia
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA; Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA; Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
7
|
Tursi NJ, Reeder SM, Flores-Garcia Y, Bah MA, Mathis-Torres S, Salgado-Jimenez B, Esquivel R, Xu Z, Chu JD, Humeau L, Patel A, Zavala F, Weiner DB. Engineered DNA-encoded monoclonal antibodies targeting Plasmodium falciparum circumsporozoite protein confer single dose protection in a murine malaria challenge model. Sci Rep 2022; 12:14313. [PMID: 35995959 PMCID: PMC9395511 DOI: 10.1038/s41598-022-18375-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/06/2022] [Indexed: 12/15/2022] Open
Abstract
Novel approaches for malaria prophylaxis remain important. Synthetic DNA-encoded monoclonal antibodies (DMAbs) are a promising approach to generate rapid, direct in vivo host-generated mAbs with potential benefits in production simplicity and distribution coupled with genetic engineering. Here, we explore this approach in a malaria challenge model. We engineered germline-reverted DMAbs based on human mAb clones CIS43, 317, and L9 which target a junctional epitope, major repeat, and minor repeat of the Plasmodium falciparum circumsporozoite protein (CSP) respectively. DMAb variants were encoded into a plasmid vector backbone and their expression and binding profiles were characterized. We demonstrate long-term serological expression of DMAb constructs resulting in in vivo efficacy of CIS43 GL and 317 GL in a rigorous mosquito bite mouse challenge model. Additionally, we engineered an Fc modified variant of CIS43 and L9-based DMAbs to ablate binding to C1q to test the impact of complement-dependent Fc function on challenge outcomes. Complement knockout variant DMAbs demonstrated similar protection to that of WT Fc DMAbs supporting the notion that direct binding to the parasite is sufficient for the protection observed. Further investigation of DMAbs for malaria prophylaxis appears of importance.
Collapse
Affiliation(s)
- Nicholas J Tursi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sophia M Reeder
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mamadou A Bah
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Shamika Mathis-Torres
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Berenice Salgado-Jimenez
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Rianne Esquivel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Ziyang Xu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jacqueline D Chu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Laurent Humeau
- Inovio Pharmaceuticals, Plymouth Meeting, PA, 19462, USA
| | - Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
8
|
Singh SK, Kumar D, Nagpal S, Dubey SK, Rathore AS. A Charge Variant of Bevacizumab Offers Enhanced FcRn-Dependent Pharmacokinetic Half-Life and Efficacy. Pharm Res 2022; 39:851-865. [PMID: 35355206 DOI: 10.1007/s11095-022-03236-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/14/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Lysine variants of monoclonal antibodies (mAbs) result from incomplete clipping of the C-terminal lysine residues of the heavy chain. Although the structure of the lysine variants has been determined for several mAb products, a detailed study that investigates the impact of lysine charge variants on PK/PD and preclinical safety is yet to be published. OBJECTIVE An in-depth investigation of the impact of C- terminal lysine clipping of mAbs on safety and efficacy for bevacizumab charge variants. METHOD Charge variant isolation using semi-preparative chromatography is followed by a comparative analysis of FcRn binding, pharmacokinetics, and pharmacodynamics in relevant animal models. RESULTS K1 variant exhibited improved FcRn binding affinity (4-fold), half-life (1.3-fold), and anti-tumor activity (1.3-fold) as compared to the K0 (main) product. However, the K2 variant, even though exhibited higher FcRn affinity (2-fold), displayed lower half-life (1.6-fold) and anti-tumor activity at medium and low doses. Differential proteomic analysis revealed that seven pathways (such as glycolysis, gluconeogenesis, carbon metabolism, synthesis of amino acids) were significantly enriched. Higher efficacy of the K1 variant is likely due to higher bioavailability of the drug, leading to complete downregulation of the pathways that facilitate catering of the energy requirements of the proliferating tumor cells. On the contrary, the K2 variant exhibits a shorter half-life, resulting only in partial reduction in the metabolic/energy requirements of the growing tumor cells. CONCLUSION Overall, we conclude that the mAb half-life, dosage, and efficacy of a biotherapeutic product are significantly impacted by the charge variant profile of a biotherapeutic product.
Collapse
Affiliation(s)
- Sumit K Singh
- School of Biochemical Engineering, IIT(BHU), Varanasi, India
| | - Deepak Kumar
- Department of Chemical Engineering, IIT, DBT Center of Excellence for Biopharmaceutical Technology, Indian Institute of Technology, Hauz Khas, New Delhi, 110016, India
| | | | - Sunil K Dubey
- R&D Healthcare Division, Emami Limited, Kolkata, India
| | - Anurag S Rathore
- Department of Chemical Engineering, IIT, DBT Center of Excellence for Biopharmaceutical Technology, Indian Institute of Technology, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
9
|
Nie W, Yao Y, Luo B, Zhu J, Li S, Yang X, Luo T, Liu W, Yan S. Systematic Analysis of the Expression and Prognosis of Fcγ Receptors in Clear Cell Renal Cell Carcinoma. Front Oncol 2022; 12:755936. [PMID: 35372055 PMCID: PMC8969749 DOI: 10.3389/fonc.2022.755936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 02/17/2022] [Indexed: 01/08/2023] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) remains a common malignancy in the urinary system. Although dramatic progress was made in multimodal therapies, the improvement of its prognosis continues to be unsatisfactory. The antibody-binding crystallizable fragment (Fc) γ receptors (FcγRs) are expressed on the surface of leukocytes, to mediate antibody-induced cell-mediated anti-tumor responses when tumor-reactive antibodies are present. FcγRs have been studied extensively in immune cells, but rarely in cancer cells. Methods ONCOMINE, UALCAN, GEPIA, TIMER, TISIDB, Kaplan–Meier Plotter, SurvivalMeth, and STRING databases were utilized in this study. Results Transcriptional levels of FcγRs were upregulated in patients with ccRCC. There was a noticeable correlation between the over expressions of FCGR1A/B/C, FCGR2A, and clinical cancer stages/tumor grade in ccRCC patients. Besides, higher transcription levels of FcγRs were found to be associated with poor overall survival (OS) in ccRCC patients. Further, high DNA methylation levels of FcγRs were also observed in ccRCC patients, and higher DNA methylation levels of FcγRs were associated with shorter OS. Moreover, we also found that the expression of FcγRs was significantly correlated with immune infiltrates, namely, immune cells (NK, macrophages, Treg, cells) and immunoinhibitor (IL-10, TGFB1, and CTLA-4). Conclusions Our study demonstrated that high DNA methylation levels of FcγRs lead to their low mRNA, protein levels, and poor prognosis in ccRCC patients, which may provide new insights into the choice of immunotherapy targets and prognostic biomarkers.
Collapse
|
10
|
Osinski V, Srikakulapu P, Haider YM, Marshall MA, Ganta VC, Annex BH, McNamara CA. Loss of Id3 (Inhibitor of Differentiation 3) Increases the Number of IgM-Producing B-1b Cells in Ischemic Skeletal Muscle Impairing Blood Flow Recovery During Hindlimb Ischemia. Arterioscler Thromb Vasc Biol 2022; 42:6-18. [PMID: 34809449 PMCID: PMC8702457 DOI: 10.1161/atvbaha.120.315501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Neovascularization can maintain and even improve tissue perfusion in the setting of limb ischemia during peripheral artery disease. The molecular and cellular mechanisms mediating this process are incompletely understood. We investigate the potential role(s) for Id3 (inhibitor of differentiation 3) in regulating blood flow in a murine model of hindlimb ischemia (HLI). Approach and Results: HLI was modeled through femoral artery ligation and resection and blood flow recovery was quantified by laser Doppler perfusion imaging. Mice with global Id3 deletion had significantly impaired perfusion recovery at 14 and 21 days of HLI. Endothelial- or myeloid cell-specific deletion of Id3 revealed no effect on perfusion recovery while B-cell-specific knockout of Id3 (Id3BKO) revealed a significant attenuation of perfusion recovery. Flow cytometry revealed no differences in ischemia-induced T cells or myeloid cell numbers at 7 days of HLI, yet there was a significant increase in B-1b cells in Id3BKO. Consistent with these findings, ELISA (enzyme-linked immunoassay) demonstrated increases in skeletal muscle and plasma IgM. In vitro experiments demonstrated reduced proliferation and increased cell death when endothelial cells were treated with conditioned media from IgM-producing B-1b cells and tibialis anterior muscles in Id3BKO mice showed reduced density of total CD31+ and αSMA+CD31+ vessels. CONCLUSIONS This study is the first to demonstrate a role for B-cell-specific Id3 in maintaining blood flow recovery during HLI. Results suggest a role for Id3 in promoting blood flow during HLI and limiting IgM-expressing B-1b cell expansion. These findings present new mechanisms to investigate in peripheral artery disease pathogenesis.
Collapse
Affiliation(s)
- Victoria Osinski
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia 22908
- Department of Pathology, University of Virginia, Charlottesville, Virginia 22908
| | - Prasad Srikakulapu
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22908
| | - Young Min Haider
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22908
| | - Melissa A. Marshall
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22908
| | - Vijay C. Ganta
- Vascular Biology Center, Augusta University, Augusta, Georgia 30912
| | - Brian H. Annex
- Vascular Biology Center, Augusta University, Augusta, Georgia 30912
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia 30912
| | - Coleen A. McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia 22908
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22908
- Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia 22908
| |
Collapse
|
11
|
Heparin-binding VEGFR1 variants as long-acting VEGF inhibitors for treatment of intraocular neovascular disorders. Proc Natl Acad Sci U S A 2021; 118:1921252118. [PMID: 34006633 PMCID: PMC8166142 DOI: 10.1073/pnas.1921252118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neovascularization is a key feature of ischemic retinal diseases and the wet form of age-related macular degeneration (AMD), all leading causes of severe vision loss. Vascular endothelial growth factor (VEGF) inhibitors have transformed the treatment of these disorders. Millions of patients have been treated with these drugs worldwide. However, in real-life clinical settings, many patients do not experience the same degree of benefit observed in clinical trials, in part because they receive fewer anti-VEGF injections. Therefore, there is an urgent need to discover and identify novel long-acting VEGF inhibitors. We hypothesized that binding to heparan-sulfate proteoglycans (HSPG) in the vitreous, and possibly other ocular structures, may be a strategy to promote intraocular retention, ultimately leading to a reduced burden of intravitreal injections. We designed a series of VEGF receptor 1 variants and identified some with strong heparin-binding characteristics and ability to bind to vitreous matrix. Our data indicate that some of our variants have longer duration and greater efficacy in animal models of intraocular neovascularization than current standard of care. Our study represents a systematic attempt to exploit the functional diversity associated with heparin affinity of a VEGF receptor.
Collapse
|
12
|
Cellular Fitness Phenotypes of Cancer Target Genes from Oncobiology to Cancer Therapeutics. Cells 2021; 10:cells10020433. [PMID: 33670680 PMCID: PMC7921985 DOI: 10.3390/cells10020433] [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: 01/15/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/17/2022] Open
Abstract
To define the growing significance of cellular targets and/or effectors of cancer drugs, we examined the fitness dependency of cellular targets and effectors of cancer drug targets across human cancer cells from 19 cancer types. We observed that the deletion of 35 out of 47 cellular effectors and/or targets of oncology drugs did not result in the expected loss of cell fitness in appropriate cancer types for which drugs targeting or utilizing these molecules for their actions were approved. Additionally, our analysis recognized 43 cellular molecules as fitness genes in several cancer types in which these drugs were not approved, and thus, providing clues for repurposing certain approved oncology drugs in such cancer types. For example, we found a widespread upregulation and fitness dependency of several components of the mevalonate and purine biosynthesis pathways (currently targeted by bisphosphonates, statins, and pemetrexed in certain cancers) and an association between the overexpression of these molecules and reduction in the overall survival duration of patients with breast and other hard-to-treat cancers, for which such drugs are not approved. In brief, the present analysis raised cautions about off-target and undesirable effects of certain oncology drugs in a subset of cancers where the intended cellular effectors of drug might not be good fitness genes and that this study offers a potential rationale for repurposing certain approved oncology drugs for targeted therapeutics in additional cancer types.
Collapse
|
13
|
Xue B, Zhang S, Gan L, Lu W, Li J. A new hand-held holder optimizes the parameters of the laser-induced choroidal neovascularization model in mice. Exp Eye Res 2020; 203:108392. [PMID: 33338490 DOI: 10.1016/j.exer.2020.108392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/09/2020] [Accepted: 12/05/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND The laser-induced choroidal neovascularization (CNV) mouse model, as the most classic animal model of age-related macular degeneration (AMD), has been widely used. We designed a hand-held mouse holder to optimize mouse fixation in the laser-induced CNV modelling process, which was inconvenient until now. This study aimed to evaluate the effectiveness of our in-house hand-held mouse holder design in the laser-induced CNV mouse modelling process. METHODS Six ophthalmic residents were invited to perform laser-induced CNV mouse modelling by hand or using the holder. We compared the learning time of residents and their physical and mental fatigue with the two methods. In addition, we compared the parameters of CNV modelling with two methods by a skilled operator, including the time of photocoagulation, induction rate and uniformity of CNV lesions. RESULTS In the learning phase, the average learning time to master the modelling method was significantly shortened by utilizing the holder. The fatigue in the operation process was quantified to a level from 0 to 4, and the physical fatigue by using holder (0.8 ± 0.3) was lower than by hand (2.6 ± 0.4), and the mental fatigue was relieved from 2.3 ± 0.5 to 0.4 ± 0.3. On the other hand, the skilled operator can significantly shorten the time of laser photocoagulation from 146.7 ± 36.0 s to 63.6 + 5.7 s and improve the success rate of modelling from 50.0% ± 8.3%-87.5% ± 6.7% by using a holder compared to hand. In addition, the standard error of the mean (SEM) of the distance between the CNV lesion and the optic nerve (ON) and the distance between each lesion was reduced. CONCLUSION This hand-held mouse holder could optimize the setting and conditions of laser-induced CNV mouse modelling by improving the learning curve, reducing fatigue, shortening the time for photocoagulation, improving the success rate and consistency of laser-induced lesions.
Collapse
Affiliation(s)
- Bai Xue
- Institute of Laboratory Medicine, Sichuan Provincial Key Laboratory for Human, Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Shanshan Zhang
- Institute of Laboratory Medicine, Sichuan Provincial Key Laboratory for Human, Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Li Gan
- Institute of Laboratory Medicine, Sichuan Provincial Key Laboratory for Human, Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Weifeng Lu
- Anesthesia Operation Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jie Li
- Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
14
|
Systemic capillary extravasation syndrome (Clarkson's syndrome) in a patient with primary hypothyroidism. Med Clin (Barc) 2020; 157:213-214. [PMID: 32843222 DOI: 10.1016/j.medcli.2020.05.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 11/22/2022]
|
15
|
Zhang L, Deng Y, Zhang Y, Liu C, Zhang S, Zhu W, Tang Y, Deng N. The Design, Characterizations, and Tumor Angiogenesis Inhibition of a Multi-Epitope Peptibody With bFGF/VEGFA. Front Oncol 2020; 10:1190. [PMID: 32766160 PMCID: PMC7379876 DOI: 10.3389/fonc.2020.01190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor angiogenesis is dependent on growth factors, and inhibition of their pathways is one of the promising strategies in cancer therapy. However, resistance to single pathway has been a great concern in clinical trials so that it necessitates multiple targetable factors for developing tumor angiogenesis inhibitors. Moreover, the strategy of Fc fusion protein is an attractive platform for novel peptide agents, which gains increasing importance with FDA approval because of better immunogenicity and stability. Here, we applied the Fc fusion protein concept to bFGF/VEGFA pathways and designed a multi-epitope Peptibody with immunogenic peptides derived from human bFGF and VEGFA sequences. Immunization with Peptibody could elicit high-titer anti-bFGF and anti-VEGFA antibodies, activate T cells, and induce Th1/Th2-type cytokines. In in vitro experiments, the isolated anti-Peptibody antibody inhibited the proliferation and migration of A549 cells and human umbilical vein endothelial cells (HUVECs) by decreasing the MAPK/Akt/mTOR signal pathways. In the murine tumor model, pre-immunization with Peptibody suppressed the tumor growth and neovascularization of lung cancer by decreasing the production of bFGF/VEGFA/PDGF, the MAPK/Akt/mTOR signal pathways, and the activation of suppressive cells in tumor sites. Further, the biological characterizations of the recombinant Peptibody were investigated systematically, including protein primary structure, secondary structure, stability, and toxicity. Collectively, the results highlighted the strategy of bFGF/VEGFA pathways and Fc fusion protein in suppressing tumor progression and angiogenesis, which emphasized the potential of multiple targetable factors for producing enduring clinical responses in tumor patients.
Collapse
Affiliation(s)
- Ligang Zhang
- Department of Biology, Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, Guangzhou, China
| | - Yanrui Deng
- Department of Biology, Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, Guangzhou, China
| | - Yinmei Zhang
- Department of Biology, Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, Guangzhou, China
| | - Chunyan Liu
- Department of Biology, Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, Guangzhou, China
| | - Simin Zhang
- Department of Biology, Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, Guangzhou, China
| | - Wenhui Zhu
- Department of Biology, Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, Guangzhou, China
| | - Yong Tang
- Department of Biology, Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, Guangzhou, China
| | - Ning Deng
- Department of Biology, Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, Guangzhou, China
| |
Collapse
|
16
|
Zhao Z, Li J, Li H, Yuan Wu NY, Ou-Yang P, Liu S, Cai J, Wang J. Integrative Bioinformatics Approaches to Screen Potential Prognostic Immune-Related Genes and Drugs in the Cervical Cancer Microenvironment. Front Genet 2020; 11:727. [PMID: 32733542 PMCID: PMC7359727 DOI: 10.3389/fgene.2020.00727] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/15/2020] [Indexed: 12/23/2022] Open
Abstract
In developing countries, cervical cancer is still the major cause of cancer-related death among women. To better understand the correlation between tumor microenvironment (TME) and prognosis of cervical cancer, we screened 1367 differentially expressed genes (DEGs) of cervical cancer samples in The Cancer Genome Atlas (TCGA) database using Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) algorithm-derived immune scores. Then, we extracted 401 tumor immune microenvironment (TIME)-related DEGs that related to patients' survival outcomes. Protein-protein interaction (PPI) network and functional enrichment analysis revealed that the prognostic genes mainly participated in myeloid leukocyte activation, adaptive immune response regulation, and receptor signaling pathways. A total of 79 key prognostic DEGs were obtained through PPI network. A TF-lncRNA-miRNA-mRNA regulatory network was constructed to explore the potential regulatory mechanism. 4 genes (CCR7, PD-1, ZAP70, and CD28) were validated in another independent cohort of cervical cancer from the Gene Expression Omnibus (GEO) database. Finally, potential drugs for key prognostics DEGs were predicted using DrugBank. In conclusion, we obtained a list of potential prognostic TIME-related genes and potential predicted drugs by integrative bioinformatics approaches. A comprehensive understanding of prognostic genes within the TIME may provide new strategies for cervical cancer treatment.
Collapse
Affiliation(s)
- Zitong Zhao
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Medicine, University of South China, Hengyang, China
| | - Jigang Li
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - He Li
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Na-Yi Yuan Wu
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Peilin Ou-Yang
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shan Liu
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jingting Cai
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jing Wang
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
17
|
de Aguiar RB, de Moraes JZ. Exploring the Immunological Mechanisms Underlying the Anti-vascular Endothelial Growth Factor Activity in Tumors. Front Immunol 2019; 10:1023. [PMID: 31156623 PMCID: PMC6530399 DOI: 10.3389/fimmu.2019.01023] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 04/23/2019] [Indexed: 12/14/2022] Open
Abstract
Several studies report the key role of the vascular endothelial growth factor (VEGF) signaling on angiogenesis and on tumor growth. This has led to the development of a number of VEGF-targeted agents to treat cancer patients by disrupting the tumor blood vessel supply. Of them, bevacizumab, an FDA-approved humanized monoclonal antibody against VEGF, is the most promising. Although the use of antibodies targeting the VEGF pathway has shown clinical benefits associated with a reduction in the tumor blood vessel density, the inhibition of VEGF-driven vascular effects is only part of the functional mechanism of these therapeutic agents in the tumor ecosystem. Compelling reports have demonstrated that VEGF confers, in addition to the activation of angiogenesis-related processes, immunosuppressive properties in tumors. It is also known that structural remodeling of the tumor blood vessel bed by anti-VEGF approaches affect the influx and activation of immune cells into tumors, which might influence the therapeutic results. Besides that, part of the therapeutic effects of antiangiogenic antibodies, including their role in the tumor vascular network, might be triggered by Fc receptors in an antigen-independent manner. In this mini-review, we explore the role of VEGF inhibitors in the tumor microenvironment with focus on the immune system, discussing around the functional contribution of both bevacizumab's Fab and Fc domains to the therapeutic results and the combination of bevacizumab therapy with other immune-stimulatory settings, including adjuvant-based vaccine approaches.
Collapse
|
18
|
Du J, Li J. The role of Wnt signaling pathway in atherosclerosis and its relationship with angiogenesis. Exp Ther Med 2018; 16:1975-1981. [PMID: 30186427 PMCID: PMC6122298 DOI: 10.3892/etm.2018.6397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/21/2018] [Indexed: 12/20/2022] Open
Abstract
Expression and function of Wnt signaling pathway in rats with atherosclerosis (AS) were investegated. The AS model of rats was established after 8-week continuous feeding of a high-fat diet, with normal rats as the control. Blood was taken from the carotid artery to detect the level of blood lipid. Aortic slices were made to observe the pathological changes of the aorta after the rats were sacrificed. Enzyme-linked immunosorbent assay kits were used to detect the contents of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Western blot analysis and semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR) were performed to detect the expression levels of related proteins and mRNA in rat Wnt signaling pathway. Correlation analysis between the protein expression level of vascular endothelial growth factor (VEGF) and that of Wnt1 was conducted. Aortic slices showed that the ratio of intima thickness to media thickness of the rats in the model group was higher than that of in the control group (P<0.01). Blood lipid and the contents of IL-6 and TNF-α of the rats in the model group were higher than those of the rats in the control group (P<0.01). Semi-quantitative RT-PCR indicated that mRNA expression levels of Wnt1, β-catenin and dickkopf1 in the aorta of rats in the model group were increased compared with those of control group (P<0.01). The results of western blot analysis revealed that the protein expression levels of Wnt1, β-catenin, DKK1 and VEGF of the rats in the model group were remarkably higher than those of the control group (P<0.01). The level of VEGF protein was positively correlated with that of Wnt1 (P<0.05, r=0.7810). The activation of Wnt signaling pathway in the aorta of the rats with AS can induce the expression of relevant inflammatory cytokines. It has the effects of promoting the progression of AS and accelerating angiogenesis.
Collapse
Affiliation(s)
- Jingru Du
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Junfeng Li
- Department of Orthopedics, The 180th Hospital of PLA (Affiliated Haixia Hospital of Huaqiao University), Quanzhou, Fujian 362008, P.R. China
| |
Collapse
|
19
|
Yerramothu P. New Therapies of Neovascular AMD-Beyond Anti-VEGFs. Vision (Basel) 2018; 2:vision2030031. [PMID: 31735894 PMCID: PMC6835305 DOI: 10.3390/vision2030031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/24/2018] [Accepted: 07/27/2018] [Indexed: 12/29/2022] Open
Abstract
Neovascular age-related macular degeneration (nAMD) is one of the leading causes of blindness among the aging population. The current treatment options for nAMD include intravitreal injections of anti-vascular endothelial growth factor (anti-VEGF). However, standardized frequent administration of anti-VEGF injections only improves vision in approximately 30–40% of nAMD patients. Current therapies targeting nAMD pose a significant risk of retinal fibrosis and geographic atrophy (GA) development in nAMD patients. A need exists to develop new therapies to treat nAMD with effective and long-term anti-angiogenic effects. Recent research on nAMD has identified novel therapeutic targets and angiogenic signaling mechanisms involved in its pathogenesis. For example, tissue factor, human intravenous immune globulin, interferon-β signaling, cyclooxygenase-2 (COX-2) and cytochrome P450 monooxygenase lipid metabolites have been identified as key players in the development of angiogenesis in AMD disease models. Furthermore, novel therapies such as NACHT, LRR and PYD domains containing protein 3 (NLRP3) inflammasome inhibition, inhibitors of integrins and tissue factor are currently being tested at the level of clinical trials to treat nAMD. The aim of this review is to discuss the scope for alternative therapies proposed as anti-VEGFs for the treatment of nAMD.
Collapse
Affiliation(s)
- Praveen Yerramothu
- School of Optometry and Vision Science, University of New South Wales, Sydney 00098, Australia
| |
Collapse
|
20
|
Apicella I, Cicatiello V, Acampora D, Tarallo V, De Falco S. Full Functional Knockout of Placental Growth Factor by Knockin with an Inactive Variant Able to Heterodimerize with VEGF-A. Cell Rep 2018; 23:3635-3646. [DOI: 10.1016/j.celrep.2018.05.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/17/2018] [Accepted: 05/18/2018] [Indexed: 12/18/2022] Open
|
21
|
Dai L, Pan Q, Peng Y, Huang S, Liu J, Chen T, Wang X, Chen D, Wang J, Zhu Y, Wang H, Liu Y, Ou Y, Yu X, Cao K. p53 Plays a Key Role in the Apoptosis of Human Ovarian Cancer Cells Induced by Adenovirus-Mediated CRM197. Hum Gene Ther 2018; 29:916-926. [PMID: 29620944 DOI: 10.1089/hum.2017.186] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cross-reacting material 197 (CRM197) is a mutant form of the diphtheria toxin. Recent studies have found that CRM197 exerts an experimental antitumor effect on several types of tumors. This study applied a novel treatment of adenovirus-mediated CRM197 (AdCRM197) to human ovarian cancer cells. Interestingly, it was found that A2780 cells were sensitive to AdCRM197, but SKOV3 cells were resistant to it. Since SKOV3 cells are p53 deletion cells, while A2780 cells are p53 wild-type cells, it was postulated that p53 might play a key role in AdCRM197-induced apoptosis. This presumption was demonstrated by means of knockdown of p53 of the A2780 cells through lentivirus-mediated RNA interference. This knockdown resulted in the A2780 cells becoming resistant to AdCRM197. To verify this presumption further, the wild-type p53 gene in the SKOV3 cells was replaced with adenovirus-mediated p53 (Adp53). As expected, AdCRM197 plus Adp53 resulted in apoptosis of the SKOV3 cells. The combined treatment of AdCRM197 plus Adp53 also showed a good antitumor effect in the in vivo experiment on nude mice with xenograft tumors. Taking these results together, it is concluded that AdCRM197 induces apoptosis of human ovarian cancer cells via the p53 pathway. Moreover, it was found that Adp53 can reverse the resistance of p53-deletion human ovarian cancer cells to AdCRM197. The combination of AdCRM197 and Adp53 may be a potentially effective method for overcoming the resistance of p53-deficient human ovarian cancer to AdCRM197.
Collapse
Affiliation(s)
- Lvxia Dai
- 1 Experiment Teaching Center of Clinical Medicine, Chengdu Medical College , Chengdu, China
| | - Qu Pan
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Yanjuan Peng
- 3 Department of Pharmacology, Chengdu Medical College , Chengdu, China
| | - Sizhou Huang
- 4 Development and Regeneration Key Laboratory of Sichuan Province, Chengdu Medical College , Chengdu, China
| | - Jianmin Liu
- 5 Department of Neurosurgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine , Guangzhou, China
| | - Tian Chen
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Xin Wang
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Dengbang Chen
- 1 Experiment Teaching Center of Clinical Medicine, Chengdu Medical College , Chengdu, China
| | - Jiandong Wang
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Yanfeng Zhu
- 6 School of Public Health, Chengdu Medical College , Chengdu, China
| | - Hui Wang
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Yilun Liu
- 7 Center for Scientific Research , First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Yu Ou
- 6 School of Public Health, Chengdu Medical College , Chengdu, China
| | - Xiaoping Yu
- 6 School of Public Health, Chengdu Medical College , Chengdu, China
| | - Kang Cao
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| |
Collapse
|
22
|
Serrano-Candelas E, Martínez-Aranguren R, Vega O, Gastaminza G, Bartra J, Audicana MT, Núñez-Córdoba JM, Algorta J, Valero A, Martin M, Ferrer M. Omalizumab efficacy in cases of chronic spontaneous urticaria is not explained by the inhibition of sera activity in effector cells. Sci Rep 2017; 7:8985. [PMID: 28827590 PMCID: PMC5566209 DOI: 10.1038/s41598-017-09361-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/24/2017] [Indexed: 11/09/2022] Open
Abstract
Omalizumab (OmAb) is a humanized anti-IgE antibody approved for the treatment of chronic spontaneous urticaria (CSU). OmAb's mechanism of action is known to include actions on free IgE and on pre-bound IgE. However, OmAb is equally and rapidly effective against autoimmune and non-autoimmune urticaria where IgE involvement is not clear, suggesting the involvement of additional mechanisms of action. In this study, we sought to investigate the ability of OmAb to inhibit mast cell and basophil degranulation induced by sera from CSU patients. For this purpose, we performed a comparison between the in vitro incubation of sera from CSU patients treated with OmAb and the in vivo administration of OmAb in a clinical trial. We found that OmAb added in vitro to sera from CSU patients did not modify the ability of the sera to induce cell degranulation. Similarly, the sera from patients treated with OmAb in the context of the clinical trial who had a good clinical outcome maintained the capacity to activate mast cells and basophils. Thus, we conclude that the beneficial activity of OmAb does not correlate with the ability of patient sera to induce cell degranulation.
Collapse
Affiliation(s)
- Eva Serrano-Candelas
- Biochemistry Unit, Faculty of Medicine, University of Barcelona, Casanova 143, Barcelona, 08036, Spain.,Laboratory of Clinical and Experimental Respiratory Immunoallergy, IDIBAPS, Barcelona, Spain
| | | | - Olga Vega
- Department of Allergy and Clinical Immunology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Gabriel Gastaminza
- Department of Allergy and Clinical Immunology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Joan Bartra
- Department of Pneumology and Allergology, Immunoallèrgia Respiratòria Clínical Experimental, IDIBAPS, Hospital Clínic, Barcelona, Spain
| | | | - Jorge M Núñez-Córdoba
- Research Support Service, Central Clinical Trials Unit, Clinica Universidad de Navarra, Pamplona, Spain
| | - Jaime Algorta
- Department of Biochemistry and Molecular Biology, Universidad del Pais Vasco-EHU, Bizkaia, Spain
| | - Antonio Valero
- Department of Pneumology and Allergology, Immunoallèrgia Respiratòria Clínical Experimental, IDIBAPS, Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias [Biomedical Research Networking Centre on Respiratory Diseases (CIBERES)], Madrid, Spain
| | - Margarita Martin
- Biochemistry Unit, Faculty of Medicine, University of Barcelona, Casanova 143, Barcelona, 08036, Spain. .,Laboratory of Clinical and Experimental Respiratory Immunoallergy, IDIBAPS, Barcelona, Spain.
| | - Marta Ferrer
- Department of Allergy and Clinical Immunology, Clinica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
23
|
Parma L, Baganha F, Quax PHA, de Vries MR. Plaque angiogenesis and intraplaque hemorrhage in atherosclerosis. Eur J Pharmacol 2017; 816:107-115. [PMID: 28435093 DOI: 10.1016/j.ejphar.2017.04.028] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/31/2017] [Accepted: 04/20/2017] [Indexed: 12/15/2022]
Abstract
Acute cardiovascular events, due to rupture or erosion of an atherosclerotic plaque, represent the major cause of morbidity and mortality in patients. Growing evidence suggests that plaque neovascularization is an important contributor to plaque growth and instability. The vessels' immaturity, with profound structural and functional abnormalities, leads to recurrent intraplaque hemorrhage. This review discusses new insights of atherosclerotic neovascularization, including the effects of leaky neovessels on intraplaque hemorrhage, both in experimental models and humans. Furthermore, modalities for in vivo imaging and therapeutic interventions to target plaque angiogenesis will be discussed.
Collapse
Affiliation(s)
- Laura Parma
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | - Fabiana Baganha
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | - Paul H A Quax
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | - Margreet R de Vries
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
24
|
Cao Z, Scandura JM, Inghirami GG, Shido K, Ding BS, Rafii S. Molecular Checkpoint Decisions Made by Subverted Vascular Niche Transform Indolent Tumor Cells into Chemoresistant Cancer Stem Cells. Cancer Cell 2017; 31:110-126. [PMID: 27989801 PMCID: PMC5497495 DOI: 10.1016/j.ccell.2016.11.010] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 04/10/2016] [Accepted: 11/17/2016] [Indexed: 02/08/2023]
Abstract
Tumor-associated endothelial cells (TECs) regulate tumor cell aggressiveness. However, the core mechanism by which TECs confer stem cell-like activity to indolent tumors is unknown. Here, we used in vivo murine and human tumor models to identify the tumor-suppressive checkpoint role of TEC-expressed insulin growth factor (IGF) binding protein-7 (IGFBP7/angiomodulin). During tumorigenesis, IGFBP7 blocks IGF1 and inhibits expansion and aggresiveness of tumor stem-like cells (TSCs) expressing IGF1 receptor (IGF1R). However, chemotherapy triggers TECs to suppress IGFBP7, and this stimulates IGF1R+ TSCs to express FGF4, inducing a feedforward FGFR1-ETS2 angiocrine cascade that obviates TEC IGFBP7. Thus, loss of IGFBP7 and upregulation of IGF1 activates the FGF4-FGFR1-ETS2 pathway in TECs and converts naive tumor cells to chemoresistant TSCs, thereby facilitating their invasiveness and progression.
Collapse
Affiliation(s)
- Zhongwei Cao
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA; Laboratory of Birth Defects and Related Diseases of Women and Children, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Joseph M Scandura
- Division of Hematology-Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giorgio G Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Koji Shido
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Bi-Sen Ding
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA; Laboratory of Birth Defects and Related Diseases of Women and Children, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Shahin Rafii
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
25
|
Ding BS, Liu CH, Sun Y, Chen Y, Swendeman SL, Jung B, Chavez D, Cao Z, Christoffersen C, Nielsen LB, Schwab SR, Rafii S, Hla T. HDL activation of endothelial sphingosine-1-phosphate receptor-1 (S1P 1) promotes regeneration and suppresses fibrosis in the liver. JCI Insight 2016; 1:e87058. [PMID: 28018969 PMCID: PMC5161208 DOI: 10.1172/jci.insight.87058] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Regeneration of hepatic sinusoidal vasculature is essential for non-fibrotic liver regrowth and restoration of its metabolic capacity. However, little is known about how this specialized vascular niche is regenerated. Here we show that activation of endothelial sphingosine-1-phosphate receptor-1 (S1P1) by its natural ligand bound to HDL (HDL-S1P) induces liver regeneration and curtails fibrosis. In mice lacking HDL-S1P, liver regeneration after partial hepatectomy was impeded and associated with aberrant vascular remodeling, thrombosis and peri-sinusoidal fibrosis. Notably, this "maladaptive repair" phenotype was recapitulated in mice that lack S1P1 in the endothelium. Reciprocally, enhanced plasma levels of HDL-S1P or administration of SEW2871, a pharmacological agonist specific for S1P1 enhanced regeneration of metabolically functional vasculature and alleviated fibrosis in mouse chronic injury and cholestasis models. This study shows that natural and pharmacological ligands modulate endothelial S1P1 to stimulate liver regeneration and inhibit fibrosis, suggesting that activation of this pathway may be a novel therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Catherine H Liu
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Yue Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yutian Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Steven L Swendeman
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA.,Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Bongnam Jung
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Deebly Chavez
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Zhongwei Cao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Department of Biomedical Sciences
| | - Lars Bo Nielsen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Department of Biomedical Sciences.,Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Susan R Schwab
- Department of Pathology, Skirball Institute, New York University School of Medicine, New York, New York, USA
| | - Shahin Rafii
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Timothy Hla
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA.,Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Ji S, Ma S, Wang WJ, Huang SZ, Wang TQ, Xiang R, Hu YG, Chen Q, Li LL, Yang SY. Discovery of selective protein arginine methyltransferase 5 inhibitors and biological evaluations. Chem Biol Drug Des 2016; 89:585-598. [PMID: 27714957 DOI: 10.1111/cbdd.12881] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/21/2016] [Accepted: 09/23/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Sen Ji
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; West China Medical School; Sichuan University; Chengdu Sichuan China
| | - Shuang Ma
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; West China Medical School; Sichuan University; Chengdu Sichuan China
| | - Wen-Jing Wang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; West China Medical School; Sichuan University; Chengdu Sichuan China
| | - Shen-Zhen Huang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; West China Medical School; Sichuan University; Chengdu Sichuan China
| | - Tian-qi Wang
- Department of Clinical Medicine; School of Medicine; Nankai University; Tianjin China
| | - Rong Xiang
- Department of Clinical Medicine; School of Medicine; Nankai University; Tianjin China
| | - Yi-Guo Hu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; West China Medical School; Sichuan University; Chengdu Sichuan China
| | - Qiang Chen
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; West China Medical School; Sichuan University; Chengdu Sichuan China
| | - Lin-Li Li
- West China School of Pharmacy; Sichuan University; Chengdu Sichuan China
| | - Sheng-Yong Yang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital; West China Medical School; Sichuan University; Chengdu Sichuan China
| |
Collapse
|
27
|
Gillis CM, Zenatti PP, Mancardi DA, Beutier H, Fiette L, Macdonald LE, Murphy AJ, Celli S, Bousso P, Jönsson F, Bruhns P. In vivo effector functions of high-affinity mouse IgG receptor FcγRI in disease and therapy models. J Autoimmun 2016; 80:95-102. [PMID: 27745779 DOI: 10.1016/j.jaut.2016.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 09/19/2016] [Accepted: 09/22/2016] [Indexed: 12/21/2022]
Abstract
Two activating mouse IgG receptors (FcγRs) have the ability to bind monomeric IgG, the high-affinity mouse FcγRI and FcγRIV. Despite high circulating levels of IgG, reports using FcγRI-/- or FcγRIV-/- mice or FcγRIV-blocking antibodies implicate these receptors in IgG-induced disease severity or therapeutic Ab efficacy. From these studies, however, one cannot conclude on the effector capabilities of a given receptor, because different activating FcγRs possess redundant properties in vivo, and cooperation between FcγRs may occur, or priming phenomena. To help resolve these uncertainties, we used mice expressing only FcγRI to determine its intrinsic properties in vivo. FcγRIonly mice were sensitive to IgG-induced autoimmune thrombocytopenia and anti-CD20 and anti-tumour immunotherapy, but resistant to IgG-induced autoimmune arthritis, anaphylaxis and airway inflammation. Our results show that the in vivo roles of FcγRI are more restricted than initially reported using FcγRI-/- mice, but confirm effector capabilities for this high-affinity IgG receptor in vivo.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/therapeutic use
- Antibody Affinity
- B-Lymphocytes/immunology
- Disease Models, Animal
- Hepatectomy
- Humans
- Immunotherapy/methods
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Purpura, Thrombocytopenic, Idiopathic/immunology
- Purpura, Thrombocytopenic, Idiopathic/therapy
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
- Splenectomy
Collapse
Affiliation(s)
- Caitlin M Gillis
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM U1222, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Priscila P Zenatti
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM U1222, Paris, France
| | - David A Mancardi
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM U1222, Paris, France
| | - Héloïse Beutier
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM U1222, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Laurence Fiette
- Département Infection et Epidémiologie, Unité d'Histopathologie Humaine et Modèles Animaux, Institut Pasteur, Paris, France
| | | | | | - Susanna Celli
- Institut Pasteur, Dynamics of Immune Responses Unit, 75015 Paris, France; INSERM U1223, rue du Dr Roux, Paris, France
| | - Philippe Bousso
- Institut Pasteur, Dynamics of Immune Responses Unit, 75015 Paris, France; INSERM U1223, rue du Dr Roux, Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM U1222, Paris, France
| | - Pierre Bruhns
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM U1222, Paris, France.
| |
Collapse
|
28
|
Abstract
Human intravenous immune globulin (IVIg), a purified IgG fraction composed of ~60% IgG1 and obtained from the pooled plasma of thousands of donors, is clinically used for a wide range of diseases. The biological actions of IVIg are incompletely understood and have been attributed both to the polyclonal antibodies therein and also to their IgG (IgG) Fc regions. Recently, we demonstrated that multiple therapeutic human IgG1 antibodies suppress angiogenesis in a target-independent manner via FcγRI, a high-affinity receptor for IgG1. Here we show that IVIg possesses similar anti-angiogenic activity and inhibited blood vessel growth in five different mouse models of prevalent human diseases, namely, neovascular age-related macular degeneration, corneal neovascularization, colorectal cancer, fibrosarcoma and peripheral arterial ischemic disease. Angioinhibition was mediated by the Fc region of IVIg, required FcγRI and had similar potency in transgenic mice expressing human FcγRs. Finally, IVIg therapy administered to humans for the treatment of inflammatory or autoimmune diseases reduced kidney and muscle blood vessel densities. These data place IVIg, an agent approved by the US Food and Drug Administration, as a novel angioinhibitory drug in doses that are currently administered in the clinical setting. In addition, they raise the possibility of an unintended effect of IVIg on blood vessels.
Collapse
|