1
|
Myo Min KK, Ffrench CB, McClure BJ, Ortiz M, Dorward EL, Samuel MS, Ebert LM, Mahoney MG, Bonder CS. Desmoglein-2 as a cancer modulator: friend or foe? Front Oncol 2023; 13:1327478. [PMID: 38188287 PMCID: PMC10766750 DOI: 10.3389/fonc.2023.1327478] [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: 10/25/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Desmoglein-2 (DSG2) is a calcium-binding single pass transmembrane glycoprotein and a member of the large cadherin family. Until recently, DSG2 was thought to only function as a cell adhesion protein embedded within desmosome junctions designed to enable cells to better tolerate mechanical stress. However, additional roles for DSG2 outside of desmosomes are continuing to emerge, particularly in cancer. Herein, we review the current literature on DSG2 in cancer and detail its impact on biological functions such as cell adhesion, proliferation, migration, invasion, intracellular signaling, extracellular vesicle release and vasculogenic mimicry. An increased understanding of the diverse repertoire of the biological functions of DSG2 holds promise to exploit this cell surface protein as a potential prognostic biomarker and/or target for better patient outcomes. This review explores the canonical and non-canonical functions of DSG2, as well as the context-dependent impacts of DSG2 in the realm of cancer.
Collapse
Affiliation(s)
- Kay K. Myo Min
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Charlie B. Ffrench
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Barbara J. McClure
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Michael Ortiz
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Emma L. Dorward
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Michael S. Samuel
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Basil Hetzel Institute, Queen Elizabeth Hospital, SA, Adelaide, Australia
| | - Lisa M. Ebert
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Mỹ G. Mahoney
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Claudine S. Bonder
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
2
|
Ramirez-Velez I, Belardi B. Storming the gate: New approaches for targeting the dynamic tight junction for improved drug delivery. Adv Drug Deliv Rev 2023; 199:114905. [PMID: 37271282 PMCID: PMC10999255 DOI: 10.1016/j.addr.2023.114905] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/20/2023] [Accepted: 05/29/2023] [Indexed: 06/06/2023]
Abstract
As biologics used in the clinic outpace the number of new small molecule drugs, an important challenge for their efficacy and widespread use has emerged, namely tissue penetrance. Macromolecular drugs - bulky, high-molecular weight, hydrophilic agents - exhibit low permeability across biological barriers. Epithelial and endothelial layers, for example within the gastrointestinal tract or at the blood-brain barrier, present the most significant obstacle to drug transport. Within epithelium, two subcellular structures are responsible for limiting absorption: cell membranes and intercellular tight junctions. Previously considered impenetrable to macromolecular drugs, tight junctions control paracellular flux and dictate drug transport between cells. Recent work, however, has shown tight junctions to be dynamic, anisotropic structures that can be targeted for delivery. This review aims to summarize new approaches for targeting tight junctions, both directly and indirectly, and to highlight how manipulation of tight junction interactions may help usher in a new era of precision drug delivery.
Collapse
Affiliation(s)
- Isabela Ramirez-Velez
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, United States
| | - Brian Belardi
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
3
|
Guo Y, Tang Y, Lu G, Gu J. p53 at the Crossroads between Doxorubicin-Induced Cardiotoxicity and Resistance: A Nutritional Balancing Act. Nutrients 2023; 15:nu15102259. [PMID: 37242146 DOI: 10.3390/nu15102259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/19/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Doxorubicin (DOX) is a highly effective chemotherapeutic drug, but its long-term use can cause cardiotoxicity and drug resistance. Accumulating evidence demonstrates that p53 is directly involved in DOX toxicity and resistance. One of the primary causes for DOX resistance is the mutation or inactivation of p53. Moreover, because the non-specific activation of p53 caused by DOX can kill non-cancerous cells, p53 is a popular target for reducing toxicity. However, the reduction in DOX-induced cardiotoxicity (DIC) via p53 suppression is often at odds with the antitumor advantages of p53 reactivation. Therefore, in order to increase the effectiveness of DOX, there is an urgent need to explore p53-targeted anticancer strategies owing to the complex regulatory network and polymorphisms of the p53 gene. In this review, we summarize the role and potential mechanisms of p53 in DIC and resistance. Furthermore, we focus on the advances and challenges in applying dietary nutrients, natural products, and other pharmacological strategies to overcome DOX-induced chemoresistance and cardiotoxicity. Lastly, we present potential therapeutic strategies to address key issues in order to provide new ideas for increasing the clinical use of DOX and improving its anticancer benefits.
Collapse
Affiliation(s)
- Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| | - Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
4
|
Richter M, Wang H, Lieber A. Role of Fiber Shaft Length in Tumor Targeting with Ad5/3 Vectors. Genes (Basel) 2022; 13:2056. [PMID: 36360292 PMCID: PMC9690795 DOI: 10.3390/genes13112056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/22/2022] [Accepted: 11/01/2022] [Indexed: 06/28/2024] Open
Abstract
Desmoglein 2 (DSG2) is overexpressed in many epithelial cancers and therefore represents a target receptor for oncolytic viruses, including Ad5/3-based viruses. For most Ad serotypes, the receptor-binding fiber is composed of tail, shaft, and knob domains. Here, we investigated the role of the fiber shaft in Ad5/3 tumor transduction in vitro and in human DSG2-transgenic mice carrying human DSG2high tumors. DSG2tg mice express DSG2 in a pattern similar to humans. We constructed Ad5/3L (with the "long" Ad5 shaft) and Ad5/3S (with the "short" Ad3 shaft) expressing GFP or luciferase. In in vitro studies we found that coagulation factor X, which is known to mediate undesired hepatocyte transduction of Ad5, enhances the transduction of Ad5/3(L), but not the transduction of Ad5/3(S). We therefore hypothesized that Ad5/3(S) would target DSG2high tumors while sparing the liver after intravenous injection. In vivo imaging studies for luciferase and analysis of luciferase activity in isolated organs, showed that Ad5/3(L) vectors efficiently transduced DSG2high tumors and liver but not normal epithelial tissues after intravenous injection. Ad5/3(S) showed minimal liver transduction, however it failed to transduce DSG2high tumors. Further modifications of the Ad5/3(S) capsid are required to compensate for the lower infectivity of Ad5/3(S) vectors.
Collapse
Affiliation(s)
| | | | - André Lieber
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
5
|
Bahlmann NA, Tsoukas RL, Erkens S, Wang H, Jönsson F, Aydin M, Naumova EA, Lieber A, Ehrhardt A, Zhang W. Properties of Adenovirus Vectors with Increased Affinity to DSG2 and the Potential Benefits of Oncolytic Approaches and Gene Therapy. Viruses 2022; 14:v14081835. [PMID: 36016457 PMCID: PMC9412290 DOI: 10.3390/v14081835] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 12/14/2022] Open
Abstract
Carcinomas are characterized by a widespread upregulation of intercellular junctions that create a barrier to immune response and drug therapy. Desmoglein 2 (DSG2) represents such a junction protein and serves as one adenovirus receptor. Importantly, the interaction between human adenovirus type 3 (Ad3) and DSG2 leads to the shedding of the binding domain followed by a decrease in the junction protein expression and transient tight junction opening. Junction opener 4 (JO-4), a small recombinant protein derived from the Ad3 fiber knob, was previously developed with a higher affinity to DSG2. JO-4 protein has been proven to enhance the effects of antibody therapy and chemotherapy and is now considered for clinical trials. However, the effect of the JO4 mutation in the context of a virus remains insufficiently studied. Therefore, we introduced the JO4 mutation to various adenoviral vectors to explore their infection properties. In the current experimental settings and investigated cell lines, the JO4-containing vectors showed no enhanced transduction compared with their parental vectors in DSG2-high cell lines. Moreover, in DSG2-low cell lines, the JO4 vectors presented a rather weakened effect. Interestingly, DSG2-negative cell line MIA PaCa-2 even showed resistance to JO4 vector infection, possibly due to the negative effect of JO4 mutation on the usage of another Ad3 receptor: CD46. Together, our observations suggest that the JO4 vectors may have an advantage to prevent CD46-mediated sequestration, thereby achieving DSG2-specific transduction.
Collapse
Affiliation(s)
- Nora A. Bahlmann
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
| | - Raphael L. Tsoukas
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
- Department of Anesthesiology and Intensive Care Medicine, Medical Faculty, University Hospital Cologne, University of Cologne, 50923 Cologne, Germany
| | - Sebastian Erkens
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
| | - Hongjie Wang
- Division of Medical Genetics, Department of Medicine, University of Washington, Box 357720, Seattle, WA 98195, USA
| | - Franziska Jönsson
- Institute of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany
| | - Malik Aydin
- Laboratory of Experimental Pediatric Pneumology and Allergology, Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 42283 Wuppertal, Germany
| | - Ella A. Naumova
- Department of Biological and Material Sciences in Dentistry, Faculty of Health, Witten/Herdecke University, 58455 Witten, Germany
| | - André Lieber
- Division of Medical Genetics, Department of Medicine, University of Washington, Box 357720, Seattle, WA 98195, USA
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
- Correspondence: (A.E.); (W.Z.)
| | - Wenli Zhang
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
- Correspondence: (A.E.); (W.Z.)
| |
Collapse
|
6
|
Wang H, Germond A, Li C, Gil S, Kim J, Kiem HP, Lieber A. In vivo HSC transduction in rhesus macaques with an HDAd5/3+ vector targeting desmoglein 2 and transiently overexpressing cxcr4. Blood Adv 2022; 6:4360-4372. [PMID: 35679480 PMCID: PMC9636333 DOI: 10.1182/bloodadvances.2022007975] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/01/2022] [Indexed: 11/20/2022] Open
Abstract
We developed a new in vivo hematopoietic stem cell (HSC) gene therapy approach that involves only IV injections and does not require myeloablation/conditioning and HSC transplantation. In this approach, HSCs are mobilized from the bone marrow into the peripheral bloodstream and transduced with IV injected helper-dependent adenovirus (HDAd) vectors. A fraction of transduced HSCs returns to the bone marrow and persists there long term. Here, we report desmoglein 2 (DSG2) as a new receptor that can be used for in vivo HSC transduction. HDAd5/3+ vectors were developed that use DSG2 as a high-affinity attachment receptor, and in vivo HSC transduction and safety after IV injection of an HDAd5/3+ vector expressing green fluorescent protein (GFP) in granulocyte colony-stimulating factor/AMD3100 (plerixafor)-mobilized rhesus macaques were studied. Unlike previously used CD46-targeting HDAd5/35++ vectors, HDAd5/3+ virions were not sequestered by rhesus erythrocytes and therefore mediated ∼10-fold higher GFP marking rates in primitive HSCs (CD34+/CD45RA-/CD90+ cells) in the bone marrow at day 7 after vector injection. To further increase the return of in vivo transduced, mobilized HSCs to the bone marrow, we transiently expressed cxcr4 in mobilized HSCs from the HDAd5/3+ vector. In vivo transduction with an HDAd5/3+GFP/cxcr4 vector at a low dose of 0.4 × 1012 viral particles/kg resulted in up to 7% of GFP-positive CD34+/CD45RA-/CD90+ cells in the bone marrow. This transduction rate is a solid basis for in vivo base or prime editing in combination with natural or drug-induced expansion of edited HSCs. Furthermore, our study provides new insights into HSC biology and trafficking after mobilization in nonhuman primates.
Collapse
Affiliation(s)
- Hongjie Wang
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA
| | - Audrey Germond
- Washington National Primate Research Center, Seattle, WA
| | - Chang Li
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA
| | - Sucheol Gil
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA
| | - Jiho Kim
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA
- PAI Life Sciences, Seattle, WA
| | - Hans-Peter Kiem
- Stem and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine
| | - André Lieber
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| |
Collapse
|
7
|
Kim J, Li C, Wang H, Kaviraj S, Singh S, Savergave L, Raghuwanshi A, Gil S, Germond A, Baldessari A, Chen B, Roffler S, Fender P, Drescher C, Carter D, Lieber A. Translational development of a tumor junction opening technology. Sci Rep 2022; 12:7753. [PMID: 35562182 PMCID: PMC9094124 DOI: 10.1038/s41598-022-11843-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/21/2022] [Indexed: 12/21/2022] Open
Abstract
Our goal is to overcome treatment resistance in ovarian cancer patients which occurs in most cases after an initial positive response to chemotherapy. A central resistance mechanism is the maintenance of desmoglein-2 (DSG2) positive tight junctions between malignant cells that prevents drug penetration into the tumor. We have generated JO4, a recombinant protein that binds to DSG2 resulting in the transient opening of junctions in epithelial tumors. Here we present studies toward the clinical translation of c-JO4 in combination with PEGylated liposomal doxorubicin/Doxil for ovarian cancer therapy. A manufacturing process for cGMP compliant production of JO4 was developed resulting in c-JO4. GLP toxicology studies using material from this process in DSG2 transgenic mice and cynomolgus macaques showed no treatment-related toxicities after intravenous injection at doses reaching 24 mg/kg. Multiple cycles of intravenous c-JO4 plus Doxil (four cycles, 4 weeks apart, simulating the treatment regimen in the clinical trial) elicited antibodies against c-JO4 that increased with each cycle and were accompanied by elevation of pro-inflammatory cytokines IL-6 and TNFα. Pretreatment with steroids and cyclophosphamide reduced anti-c-JO4 antibody response and blunted cytokine release. Our data indicate acceptable safety of our new treatment approach if immune reactions are monitored and counteracted with appropriate immune suppression.
Collapse
Affiliation(s)
- Jiho Kim
- Division of Medical Genetics, School of Medicine, University of Washington, Seattle, WA, USA
- PAI Life Sciences, Seattle, WA, USA
| | - Chang Li
- Division of Medical Genetics, School of Medicine, University of Washington, Seattle, WA, USA
| | - Hongjie Wang
- Division of Medical Genetics, School of Medicine, University of Washington, Seattle, WA, USA
| | | | | | | | | | - Sucheol Gil
- Division of Medical Genetics, School of Medicine, University of Washington, Seattle, WA, USA
| | - Audrey Germond
- Washington National Primate Research Center, Seattle, WA, USA
| | | | - Bingmae Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Steve Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Pascal Fender
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042, Grenoble, France
| | - Charles Drescher
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Darrick Carter
- Division of Medical Genetics, School of Medicine, University of Washington, Seattle, WA, USA
- PAI Life Sciences, Seattle, WA, USA
- Department of Global Health, School of Public Health, University of Washington, Seattle, WA, USA
| | - André Lieber
- Division of Medical Genetics, School of Medicine, University of Washington, Seattle, WA, USA.
- Department of Pathology, School of Medicine, University of Washington, Box 357720, Seattle, WA, 98195, USA.
| |
Collapse
|
8
|
Targeting Cancer Cell Tight Junctions Enhances PLGA-Based Photothermal Sensitizers' Performance In Vitro and In Vivo. Pharmaceutics 2021; 14:pharmaceutics14010043. [PMID: 35056939 PMCID: PMC8778343 DOI: 10.3390/pharmaceutics14010043] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/11/2021] [Accepted: 12/22/2021] [Indexed: 12/16/2022] Open
Abstract
The development of non-invasive photothermal therapy (PTT) methods utilizing nanoparticles as sensitizers is one of the most promising directions in modern oncology. Nanoparticles loaded with photothermal dyes are capable of delivering a sufficient amount of a therapeutic substance and releasing it with the desired kinetics in vivo. However, the effectiveness of oncotherapy methods, including PTT, is often limited due to poor penetration of sensitizers into the tumor, especially into solid tumors of epithelial origin characterized by tight cellular junctions. In this work, we synthesized 200 nm nanoparticles from the biocompatible copolymer of lactic and glycolic acid, PLGA, loaded with magnesium phthalocyanine, PLGA/Pht-Mg. The PLGA/Pht-Mg particles under the irradiation with NIR light (808 nm), heat the surrounding solution by 40 °C. The effectiveness of using such particles for cancer cells elimination was demonstrated in 2D culture in vitro and in our original 3D model with multicellular spheroids possessing tight cell contacts. It was shown that the mean inhibitory concentration of such nanoparticles upon light irradiation for 15 min worsens by more than an order of magnitude: IC50 increases from 3 µg/mL for 2D culture vs. 117 µg/mL for 3D culture. However, when using the JO-4 intercellular junction opener protein, which causes a short epithelial–mesenchymal transition and transiently opens intercellular junctions in epithelial cells, the efficiency of nanoparticles in 3D culture was comparable or even outperforming that for 2D (IC50 = 1.9 µg/mL with JO-4). Synergy in the co-administration of PTT nanosensitizers and JO-4 protein was found to retain in vivo using orthotopic tumors of BALB/c mice: we demonstrated that the efficiency in the delivery of such nanoparticles to the tumor is 2.5 times increased when PLGA/Pht-Mg nanoparticles are administered together with JO-4. Thus the targeting the tumor cell junctions can significantly increase the performance of PTT nanosensitizers.
Collapse
|
9
|
Brunner J, Ragupathy S, Borchard G. Target specific tight junction modulators. Adv Drug Deliv Rev 2021; 171:266-288. [PMID: 33617902 DOI: 10.1016/j.addr.2021.02.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Intercellular tight junctions represent a formidable barrier against paracellular drug absorption at epithelia (e.g., nasal, intestinal) and the endothelium (e.g., blood-brain barrier). In order to enhance paracellular transport of drugs and increase their bioavailability and organ deposition, active excipients modulating tight junctions have been applied. First-generation of permeation enhancers (PEs) acted by unspecific interactions, while recently developed PEs address specific physiological mechanisms. Such target specific tight junction modulators (TJMs) have the advantage of a defined specific mechanism of action. To date, merely a few of these novel active excipients has entered into clinical trials, as their lack in safety and efficiency in vivo often impedes their commercialisation. A stronger focus on the development of such active excipients would result in an economic and therapeutic improvement of current and future drugs.
Collapse
Affiliation(s)
- Joël Brunner
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Sakthikumar Ragupathy
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
10
|
Kim J, Beidler P, Wang H, Li C, Quassab A, Coles C, Drescher C, Carter D, Lieber A. Desmoglein-2 as a prognostic and biomarker in ovarian cancer. Cancer Biol Ther 2020; 21:1154-1162. [PMID: 33218274 PMCID: PMC7722792 DOI: 10.1080/15384047.2020.1843323] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/31/2023] Open
Abstract
Greater than 80% of all cancer cases are carcinomas, formed by the malignant transformation of epithelial cells. One of the key features of epithelial tumors is the presence of intercellular junctions, which link cells to one another and act as barriers to the penetration of molecules. This study assessed the expression of desmoglein-2, an epithelial junction protein, as a prognostic and diagnostic biomarker for ovarian cancer. Ovarian cancer sections were stained for DSG2 and signal intensity was correlated to cancer type and grade. DSG2 immunohistochemistry signals and mRNA levels were analyzed in chemo-resistant and chemo-sensitive cases. Ovarian cancer patient serum levels of shed DSG2 were correlated to disease-free and overall survival. Primary ovarian cancer cells were used to study DSG2 levels as they changed in response to cisplatin treatment. DSG2 expression was found to be positively correlated with cancer grade. Ovarian cancer patients with high serum levels of shed DSG2 fared significantly worse in both progression-free survival (median survival of 16 months vs. 26 months, p = .0023) and general survival (median survival of 37 months vs. undefined, p < .0001). A subgroup of primary chemotherapy-resistant cases had stronger DSG2 IHC/Western signals and higher DSG2 mRNA levels. Furthermore, our in vitro studies indicate that non-cytotoxic doses of cisplatin can enhance DSG2 expression, which, in turn, can contribute to chemo-resistance. We suggest that DSG2 can be used in stratifying patients, deciding on where to use aggressive treatment strategies, predicting chemoresistance, and as a companion diagnostic for treatments targeting DSG2.
Collapse
Affiliation(s)
- Jiho Kim
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
- R&D Department, PAI Life Sciences Inc, Seattle, Washington, USA
| | - Peter Beidler
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Hongjie Wang
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Chang Li
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Abdullah Quassab
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Cari Coles
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Charles Drescher
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Darrick Carter
- R&D Department, PAI Life Sciences Inc, Seattle, Washington, USA
- R&D Department, Onc Bio, Seattle, Washington, USA
| | - André Lieber
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
Shipunova VO, Komedchikova EN, Kotelnikova PA, Zelepukin IV, Schulga AA, Proshkina GM, Shramova EI, Kutscher HL, Telegin GB, Kabashin AV, Prasad PN, Deyev SM. Dual Regioselective Targeting the Same Receptor in Nanoparticle-Mediated Combination Immuno/Chemotherapy for Enhanced Image-Guided Cancer Treatment. ACS NANO 2020; 14:12781-12795. [PMID: 32935975 DOI: 10.1021/acsnano.0c03421] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
When combined with immunotherapy, image-guided targeted delivery of chemotherapeutic agents is a promising direction for combination cancer theranostics, but this approach has so far produced only limited success due to a lack of molecular targets on the cell surface and low therapeutic index of conventional chemotherapy drugs. Here, we demonstrate a synergistic strategy of combination immuno/chemotherapy in conditions of dual regioselective targeting, implying vectoring of two distinct binding sites of a single oncomarker (here, HER2) with theranostic compounds having a different mechanism of action. We use: (i) PLGA nanoformulation, loaded with an imaging diagnostic fluorescent dye (Nile Red) and a chemotherapeutic drug (doxorubicin), and functionalized with affibody ZHER2:342 (8 kDa); (ii) bifunctional genetically engineered DARP-LoPE (42 kDa) immunotoxin comprising of a low-immunogenic modification of therapeutic Pseudomonas exotoxin A (LoPE) and a scaffold targeting protein, DARPin9.29 (14 kDa). According to the proposed strategy, the first chemotherapeutic nanoagent is targeted by the affibody to subdomain III and IV of HER2 with 60-fold specificity compared with nontargeted particles, while the second immunotoxin is effectively targeted by DARPin molecule to subdomain I of HER2. We demonstrate that this dual targeting strategy can enhance anticancer therapy of HER2-positive cells with a very strong synergy, which made possible 1000-fold decrease of effective drug concentration in vitro and a significant enhancement of HER2 cancer therapy compared to monotherapy in vivo. Moreover, this therapeutic combination prevented the appearance of secondary tumor nodes. Thus, the suggested synergistic strategy utilizing dual targeting of the same oncomarker could give rise to efficient methods for aggressive tumors treatment.
Collapse
Affiliation(s)
- Victoria O Shipunova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
- MEPhI (Moscow Engineering Physics Institute), Institute of Engineering Physics for Biomedicine (PhysBio), 31 Kashirskoe Shosse, Moscow 115409, Russia
| | - Elena N Komedchikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Polina A Kotelnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Ivan V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
- MEPhI (Moscow Engineering Physics Institute), Institute of Engineering Physics for Biomedicine (PhysBio), 31 Kashirskoe Shosse, Moscow 115409, Russia
| | - Alexey A Schulga
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Galina M Proshkina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Elena I Shramova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Hilliard L Kutscher
- Institute for Lasers, Photonics and Biophotonics, University at Buffalo, 428 Natural Science Complex, Buffalo, New York 14260-3000, United States
- Department of Medicine, University at Buffalo, 875 Ellicott Street, Buffalo, New York 14203, United States
- Department of Anesthesiology, University at Buffalo, 77 Goodell Street, Suite 550, Buffalo, New York 14203, United States
| | - Georgij B Telegin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
| | - Andrei V Kabashin
- MEPhI (Moscow Engineering Physics Institute), Institute of Engineering Physics for Biomedicine (PhysBio), 31 Kashirskoe Shosse, Moscow 115409, Russia
- Aix Marseille University, CNRS, LP3, Campus de Luminy-case 917, 13288, Marseille Cedex 9, France
| | - Paras N Prasad
- MEPhI (Moscow Engineering Physics Institute), Institute of Engineering Physics for Biomedicine (PhysBio), 31 Kashirskoe Shosse, Moscow 115409, Russia
- Institute for Lasers, Photonics and Biophotonics, University at Buffalo, 428 Natural Science Complex, Buffalo, New York 14260-3000, United States
| | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya Street, Moscow 117997, Russia
- MEPhI (Moscow Engineering Physics Institute), Institute of Engineering Physics for Biomedicine (PhysBio), 31 Kashirskoe Shosse, Moscow 115409, Russia
| |
Collapse
|
12
|
Besson S, Vragniau C, Vassal-Stermann E, Dagher MC, Fender P. The Adenovirus Dodecahedron: Beyond the Platonic Story. Viruses 2020; 12:E718. [PMID: 32630840 PMCID: PMC7412204 DOI: 10.3390/v12070718] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 02/03/2023] Open
Abstract
Many geometric forms are found in nature, some of them adhering to mathematical laws or amazing aesthetic rules. One of the best-known examples in microbiology is the icosahedral shape of certain viruses with 20 triangular facets and 12 edges. What is less known, however, is that a complementary object displaying 12 faces and 20 edges called a 'dodecahedron' can be produced in huge amounts during certain adenovirus replication cycles. The decahedron was first described more than 50 years ago in the human adenovirus (HAdV3) viral cycle. Later on, the expression of this recombinant scaffold, combined with improvements in cryo-electron microscopy, made it possible to decipher the structural determinants underlying their architecture. Recently, this particle, which mimics viral entry, was used to fish the long elusive adenovirus receptor, desmoglein-2, which serves as a cellular docking for some adenovirus serotypes. This breakthrough enabled the understanding of the physiological role played by the dodecahedral particles, showing that icosahedral and dodecahedral particles live more than a simple platonic story. All these points are developed in this review, and the potential use of the dodecahedron in therapeutic development is discussed.
Collapse
Affiliation(s)
- Solène Besson
- Centre National de la Recherche Scientifique, Université Grenoble Alpes, Commissariat Enérgies Alternatives, Institut de Biologie Structurale, 41 rue des Martyrs, 38042 Grenoble, France; (S.B.); (C.V.); (E.V.-S.); (M.C.D.)
| | - Charles Vragniau
- Centre National de la Recherche Scientifique, Université Grenoble Alpes, Commissariat Enérgies Alternatives, Institut de Biologie Structurale, 41 rue des Martyrs, 38042 Grenoble, France; (S.B.); (C.V.); (E.V.-S.); (M.C.D.)
- Centre National de la Recherche Scientifique, Université Grenoble Alpes, Institut National Polytechnique Grenoble, Technique de l’ingénierie Médicale et de la Complexité, TIMC-IMAG Bât Jean Roget Faculté de Médecine et Pharmacie, 38700 La Tronche, France
| | - Emilie Vassal-Stermann
- Centre National de la Recherche Scientifique, Université Grenoble Alpes, Commissariat Enérgies Alternatives, Institut de Biologie Structurale, 41 rue des Martyrs, 38042 Grenoble, France; (S.B.); (C.V.); (E.V.-S.); (M.C.D.)
| | - Marie Claire Dagher
- Centre National de la Recherche Scientifique, Université Grenoble Alpes, Commissariat Enérgies Alternatives, Institut de Biologie Structurale, 41 rue des Martyrs, 38042 Grenoble, France; (S.B.); (C.V.); (E.V.-S.); (M.C.D.)
| | - Pascal Fender
- Centre National de la Recherche Scientifique, Université Grenoble Alpes, Commissariat Enérgies Alternatives, Institut de Biologie Structurale, 41 rue des Martyrs, 38042 Grenoble, France; (S.B.); (C.V.); (E.V.-S.); (M.C.D.)
| |
Collapse
|
13
|
Pitner R, Kim J, Davis-Bergthold J, Turner C, Vassal-Stermann E, Wang H, Adams J, Carter L, Ahlgren JA, Fender P, Lieber A, Carter D, Gray SA. Structure-based Design of JOC-x, a Conjugatable Tumor Tight Junction Opener to Enhance Cancer Therapy. Sci Rep 2019; 9:6169. [PMID: 30992466 PMCID: PMC6467980 DOI: 10.1038/s41598-019-42229-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 03/26/2019] [Indexed: 01/14/2023] Open
Abstract
Disorganized intercellular junctions are critical for maintaining the integrity of solid epithelial tumors and prevent the infiltration of oncological therapies into the bulk of the malignancy. We have developed small, recombinant proteins which bind a critical junction protein, desmoglein 2, triggering the transient and specific opening of tumor tight junctions allowing for infiltration of the tumor with immune cells, oncolytic viruses, drugs, and other therapeutics. Our new molecule, JOC-x, is a promising candidate for a new class of tumor-targeting agents that accumulate both around and within tumors and remodel the tumor microenvironment. Native cysteines were removed from the parental protein, JO-4, followed by addition of a single cysteine to allow for convenient attachment of various payloads that can be targeted directly to the tumor. Our tumor-targeting protein exhibits high avidity, minimal aggregation, and is easily purified at good yields from E. coli. For proof of concept, we demonstrate effective conjugation to biotin as a model for flexible co-targeting, addition of metal ion chelators as models for imaging and radiotherapy, and linkage of the TLR3 agonist poly(I:C) as a model immune-oncologic agent. This second-generation cancer co-therapeutic protein is optimized for activity and primed for cGMP manufacture in preparation for upcoming clinical studies.
Collapse
Affiliation(s)
- Ragan Pitner
- grid.423437.5PAI Life Sciences, Inc., Seattle, WA USA ,0000000122986657grid.34477.33University of Washington, Dept. of Immunology, Seattle, WA USA
| | - Jiho Kim
- grid.423437.5PAI Life Sciences, Inc., Seattle, WA USA ,0000000122986657grid.34477.33University of Washington, Program in Pathobiology, Dept. of Global Health, Seattle, WA USA
| | | | - Cheri Turner
- grid.423437.5PAI Life Sciences, Inc., Seattle, WA USA
| | - Emilie Vassal-Stermann
- 0000 0004 0641 5776grid.418192.7Institut de Biologie Structurale, UMR5075, CNRS/CEA/UGA, Grenoble, France
| | - Hongjie Wang
- 0000000122986657grid.34477.33University of Washington, Division of Medical Genetics, Seattle, WA USA
| | - Jaclyn Adams
- 0000000122986657grid.34477.33University of Washington, Division of Medical Genetics, Seattle, WA USA
| | - Lauren Carter
- 0000000122986657grid.34477.33Institute for Protein Design, Department of Biochemistry, University of Washington, Seattle, WA USA
| | | | - Pascal Fender
- 0000 0004 0641 5776grid.418192.7Institut de Biologie Structurale, UMR5075, CNRS/CEA/UGA, Grenoble, France
| | - André Lieber
- 0000000122986657grid.34477.33University of Washington, Division of Medical Genetics, Seattle, WA USA ,grid.428229.7Compliment Corp., Seattle, WA USA
| | - Darrick Carter
- grid.423437.5PAI Life Sciences, Inc., Seattle, WA USA ,0000000122986657grid.34477.33University of Washington, Program in Pathobiology, Dept. of Global Health, Seattle, WA USA ,0000000122986657grid.34477.33University of Washington, Division of Medical Genetics, Seattle, WA USA ,grid.428229.7Compliment Corp., Seattle, WA USA
| | - Sean A. Gray
- grid.423437.5PAI Life Sciences, Inc., Seattle, WA USA
| |
Collapse
|
14
|
CryoEM structure of adenovirus type 3 fibre with desmoglein 2 shows an unusual mode of receptor engagement. Nat Commun 2019; 10:1181. [PMID: 30862836 PMCID: PMC6414520 DOI: 10.1038/s41467-019-09220-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/26/2019] [Indexed: 11/09/2022] Open
Abstract
Attachment of human adenovirus (HAd) to the host cell is a critical step of infection. Initial attachment occurs via the adenoviral fibre knob protein and a cellular receptor. Here we report the cryo-electron microscopy (cryo-EM) structure of a <100 kDa non-symmetrical complex comprising the trimeric HAd type 3 fibre knob (HAd3K) and human desmoglein 2 (DSG2). The structure reveals a unique stoichiometry of 1:1 and 2:1 (DSG2: knob trimer) not previously observed for other HAd-receptor complexes. We demonstrate that mutating Asp261 in the fibre knob is sufficient to totally abolish receptor binding. These data shed new light on adenovirus infection strategies and provide insights for adenoviral vector development and structure-based design.
Collapse
|
15
|
Disassembling a cancer puzzle: Cell junctions and plasma membrane as targets for anticancer therapy. J Control Release 2018; 286:125-136. [PMID: 30030181 DOI: 10.1016/j.jconrel.2018.07.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 02/07/2023]
Abstract
Despite an enhanced permeability and retention effect typical of many solid tumors, drug penetration is not always sufficient. Possible strategies for the drug delivery improvement are a modification of the tumor cell-to-cell junctions and usage of cell membrane permeabilization proteins. In this review we discuss epithelial cell junctions as targets for a combined anticancer therapy and propose new possible sources of such agents. We suggest considering viral and bacterial pathogens disrupting epithelial layers as plentiful sources of new therapeutic agents for increasing tumor permeability for other effector agents. We also observe the application of pore forming proteins and peptides of different origin for cytoplasmic delivery of anti-cancer agents and consider the main obstacles of their use in vivo.
Collapse
|
16
|
Baker AT, Aguirre-Hernández C, Halldén G, Parker AL. Designer Oncolytic Adenovirus: Coming of Age. Cancers (Basel) 2018; 10:E201. [PMID: 29904022 PMCID: PMC6025169 DOI: 10.3390/cancers10060201] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/06/2018] [Accepted: 06/11/2018] [Indexed: 12/26/2022] Open
Abstract
The licensing of talimogene laherparepvec (T-Vec) represented a landmark moment for oncolytic virotherapy, since it provided unequivocal evidence for the long-touted potential of genetically modified replicating viruses as anti-cancer agents. Whilst T-Vec is promising as a locally delivered virotherapy, especially in combination with immune-checkpoint inhibitors, the quest continues for a virus capable of specific tumour cell killing via systemic administration. One candidate is oncolytic adenovirus (Ad); it’s double stranded DNA genome is easily manipulated and a wide range of strategies and technologies have been employed to empower the vector with improved pharmacokinetics and tumour targeting ability. As well characterised clinical and experimental agents, we have detailed knowledge of adenoviruses’ mechanisms of pathogenicity, supported by detailed virological studies and in vivo interactions. In this review we highlight the strides made in the engineering of bespoke adenoviral vectors to specifically infect, replicate within, and destroy tumour cells. We discuss how mutations in genes regulating adenoviral replication after cell entry can be used to restrict replication to the tumour, and summarise how detailed knowledge of viral capsid interactions enable rational modification to eliminate native tropisms, and simultaneously promote active uptake by cancerous tissues. We argue that these designer-viruses, exploiting the viruses natural mechanisms and regulated at every level of replication, represent the ideal platforms for local overexpression of therapeutic transgenes such as immunomodulatory agents. Where T-Vec has paved the way, Ad-based vectors now follow. The era of designer oncolytic virotherapies looks decidedly as though it will soon become a reality.
Collapse
Affiliation(s)
- Alexander T Baker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - Carmen Aguirre-Hernández
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Gunnel Halldén
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Alan L Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| |
Collapse
|
17
|
Vassal-Stermann E, Mottet M, Ducournau C, Iseni F, Vragniau C, Wang H, Zubieta C, Lieber A, Fender P. Mapping of Adenovirus of serotype 3 fibre interaction to desmoglein 2 revealed a novel 'non-classical' mechanism of viral receptor engagement. Sci Rep 2018; 8:8381. [PMID: 29849084 PMCID: PMC5976663 DOI: 10.1038/s41598-018-26871-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/18/2018] [Indexed: 12/05/2022] Open
Abstract
High-affinity binding of the trimeric fibre protein to a cell surface primary receptor is a common feature shared by all adenovirus serotypes. Recently, a long elusive species B adenovirus receptor has been identified. Desmoglein 2 (DSG2) a component of desmosomal junction, has been reported to interact at high affinity with Human adenoviruses HAd3, HAd7, HAd11 and HAd14. Little is known with respect to the molecular interactions of adenovirus fibre with the DSG2 ectodomain. By using different DSG2 ectodomain constructs and biochemical and biophysical experiments, we report that the third extracellular cadherin domain (EC3) of DSG2 is critical for HAd3 fibre binding. Unexpectedly, stoichiometry studies using multi-angle laser light scattering (MALLS) and analytical ultra-centrifugation (AUC) revealed a non-classical 1:1 interaction (one DSG2 per trimeric fibre), thus differentiating ‘DSG2-interacting’ adenoviruses from other protein receptor interacting adenoviruses in their infection strategy.
Collapse
Affiliation(s)
- Emilie Vassal-Stermann
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CNRS, CEA, 71 Avenue des Martyrs, 38042, Grenoble, France
| | - Manon Mottet
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CNRS, CEA, 71 Avenue des Martyrs, 38042, Grenoble, France
| | - Corinne Ducournau
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CNRS, CEA, 71 Avenue des Martyrs, 38042, Grenoble, France.,Unité de Virologie, Institut de Recherche Biomédicale des Armées, BP 73, 91223, Brétigny-sur-Orge Cedex, France
| | - Frédéric Iseni
- Unité de Virologie, Institut de Recherche Biomédicale des Armées, BP 73, 91223, Brétigny-sur-Orge Cedex, France
| | - Charles Vragniau
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CNRS, CEA, 71 Avenue des Martyrs, 38042, Grenoble, France
| | - Hongjie Wang
- University of Washington, Department of Medicine, Division of Medical Genetics, Box 357720, Seattle, WA, 98195, USA
| | - Chloe Zubieta
- Laboratoire de Physiologie Cellulaire et Végétale, Biosciences and Biotechnology Institute of Grenoble, UMR5168, CNRS/CEA/INRA/UGA, 17 Rue des Martyrs, 38054, Grenoble, France
| | - André Lieber
- University of Washington, Department of Medicine, Division of Medical Genetics, Box 357720, Seattle, WA, 98195, USA.
| | - Pascal Fender
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CNRS, CEA, 71 Avenue des Martyrs, 38042, Grenoble, France.
| |
Collapse
|
18
|
Desmoglein 2 promotes vasculogenic mimicry in melanoma and is associated with poor clinical outcome. Oncotarget 2018; 7:46492-46508. [PMID: 27340778 PMCID: PMC5216812 DOI: 10.18632/oncotarget.10216] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/03/2016] [Indexed: 12/12/2022] Open
Abstract
Tumors can develop a blood supply not only by promoting angiogenesis but also by forming vessel-like structures directly from tumor cells, known as vasculogenic mimicry (VM). Understanding mechanisms that regulate VM is important, as these might be exploitable to inhibit tumor progression. Here, we reveal the adhesion molecule desmoglein 2 (DSG2) as a novel mediator of VM in melanoma. Analysis of patient-derived melanoma cell lines and tumor tissues, and interrogation of The Cancer Genome Atlas (TCGA) data, revealed that DSG2 is frequently overexpressed in primary and metastatic melanomas compared to normal melanocytes. Notably, this overexpression was associated with poor clinical outcome. DSG2+ melanoma cells self-organized into tube-like structures on Matrigel, indicative of VM activity, which was inhibited by DSG2 knockdown or treatment with a DSG2-blocking peptide. Mechanistic studies revealed that DSG2 regulates adhesion and cell-cell interactions during tube formation, but does not control melanoma cell viability, proliferation or motility. Finally, analysis of patient tumors revealed a correlation between DSG2 expression, VM network density and expression of VM-associated genes. These studies identify DSG2 as a key regulator of VM activity in human melanoma and suggest this molecule might be therapeutically targeted to reduce tumor blood supply and metastatic spread.
Collapse
|
19
|
Wang CE, Yumul RC, Lin J, Cheng Y, Lieber A, Pun SH. Junction opener protein increases nanoparticle accumulation in solid tumors. J Control Release 2018; 272:9-16. [PMID: 29305923 DOI: 10.1016/j.jconrel.2017.12.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/21/2017] [Accepted: 12/28/2017] [Indexed: 11/18/2022]
Abstract
Carcinomas contain tight junctions that can limit the penetration and therefore therapeutic efficacy of anticancer agents, especially those delivered by nano-carrier systems. The junction opener (JO) protein is a virus-derived protein that can transiently open intercellular junctions in epithelial tumors by cleaving the junction protein desmoglein-2 (DSG2). Co-administration of JO was previously shown to significantly increase the efficacy of various monoclonal antibodies and chemotherapy drugs in murine tumor models by allowing for increased intratumoral penetration of the drugs. To investigate the size-dependent effect of JO on nanocarriers, we used PEGylated gold nanoparticles (AuNPs) of two different sizes as model drugs and investigated their biodistribution following JO protein treatment. By inductively coupled plasma mass spectrometry (ICP-MS), JO was found to significantly increase bulk tumor accumulation of AuNPs of 35nm but not 120nm particles in both medium (200-300mm3) and large (500-600mm3) tumors. Image analysis of tumor sections corroborates this JO-mediated increase in tumor accumulation of AuNPs. Quantitative intratumoral distribution analyses show that most nanoparticles were found within 100μm of the vasculature, and that the penetration profiles of AuNPs are not significantly affected by JO treatment at the 6h timepoint.
Collapse
Affiliation(s)
- Christine E Wang
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, 3720 15th Ave NE, Seattle, WA 98195, United States
| | - Roma C Yumul
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, United States
| | - Jonathan Lin
- Department of Bioengineering, University of California, Los Angeles, CA 90095, United States
| | - Yilong Cheng
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, 3720 15th Ave NE, Seattle, WA 98195, United States
| | - André Lieber
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, United States
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, 3720 15th Ave NE, Seattle, WA 98195, United States.
| |
Collapse
|
20
|
Yumul R, Richter M, Lu ZZ, Saydaminova K, Wang H, Wang CHK, Carter D, Lieber A. Epithelial Junction Opener Improves Oncolytic Adenovirus Therapy in Mouse Tumor Models. Hum Gene Ther 2016; 27:325-37. [PMID: 26993072 DOI: 10.1089/hum.2016.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A central resistance mechanism in solid tumors is the maintenance of epithelial junctions between malignant cells that prevent drug penetration into the tumor. Human adenoviruses (Ads) have evolved mechanisms to breach epithelial barriers. For example, during Ad serotype 3 (Ad3) infection of epithelial tumor cells, massive amounts of subviral penton-dodecahedral particles (PtDd) are produced and released from infected cells to trigger the transient opening of epithelial junctions, thus facilitating lateral virus spread. We show here that an Ad3 mutant that is disabled for PtDd production is significantly less effective in killing of epithelial human xenograft tumors than the wild-type Ad3 virus. Intratumoral spread and therapeutic effect of the Ad3 mutant was enhanced by co-administration of a small recombinant protein (JO; produced in Escherichia coli) that incorporated the minimal junction opening domains of PtDd. We then demonstrated that co-administration of JO with replication-competent Ads that do not produce PtDd (Ad5, Ad35) resulted in greater attenuation of tumor growth than virus injection alone. Furthermore, we genetically modified a conditionally replicating Ad5-based oncolytic Ad (Ad5Δ24) to express a secreted form of JO upon replication in tumor cells. The JO-expressing virus had a significantly greater antitumor effect than the unmodified AdΔ24 version. Our findings indicate that epithelial junctions limit the efficacy of oncolytic Ads and that this problem can be address by co-injection or expression of JO. JO has also the potential for improving cancer therapy with other types of oncolytic viruses.
Collapse
Affiliation(s)
- Roma Yumul
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington
| | - Maximilian Richter
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington
| | - Zhuo-Zhuang Lu
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington.,2 National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing, PR China
| | - Kamola Saydaminova
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington
| | - Hongjie Wang
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington
| | | | - Darrick Carter
- 4 Compliment Corp. , Seattle, Washington.,5 PAI Life Sciences Inc. , Seattle, Washington
| | - André Lieber
- 1 Division of Medical Genetics, University of Washington , Seattle, Washington.,4 Compliment Corp. , Seattle, Washington.,6 Department of Pathology, University of Washington , Seattle, Washington
| |
Collapse
|
21
|
MMP-9 triggered self-assembly of doxorubicin nanofiber depots halts tumor growth. Biomaterials 2016; 98:192-202. [PMID: 27192421 DOI: 10.1016/j.biomaterials.2016.04.039] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/18/2016] [Accepted: 04/28/2016] [Indexed: 12/27/2022]
Abstract
A central challenge in cancer care is to ensure that therapeutic compounds reach their targets. One approach is to use enzyme-responsive biomaterials, which reconfigure in response to endogenous enzymes that are overexpressed in diseased tissues, as potential site-specific anti-tumoral therapies. Here we report peptide micelles that upon MMP-9 catalyzed hydrolysis reconfigure to form fibrillar nanostructures. These structures slowly release a doxorubicin payload at the site of action. Using both in vitro and in vivo models, we demonstrate that the fibrillar depots are formed at the sites of MMP-9 overexpression giving rise to enhanced efficacy of doxorubicin, resulting in inhibition of tumor growth in an animal model.
Collapse
|
22
|
Intracellular Signaling and Desmoglein 2 Shedding Triggered by Human Adenoviruses Ad3, Ad14, and Ad14P1. J Virol 2015; 89:10841-59. [PMID: 26292319 DOI: 10.1128/jvi.01425-15] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/10/2015] [Indexed: 01/10/2023] Open
Abstract
UNLABELLED We recently discovered that desmoglein 2 (DSG2) is a receptor for human adenovirus species B serotypes Ad3, Ad7, Ad11, and Ad14. Ad3 is considered to be a widely distributed human pathogen. Ad3 binding to DSG2 triggers the transient opening of epithelial junctions. Here, we further delineate the mechanism that leads to DSG2-mediated epithelial junction opening in cells exposed to Ad3 and recombinant Ad3 fiber proteins. We identified an Ad3 fiber knob-dependent pathway that involves the phosphorylation of mitogen-activated protein (MAP) kinases triggering the activation of the matrix-metalloproteinase ADAM17. ADAM17, in turn, cleaves the extracellular domain of DSG2 that links epithelial cells together. The shed DSG2 domain can be detected in cell culture supernatant and also in serum of mice with established human xenograft tumors. We then extended our studies to Ad14 and Ad14P1. Ad14 is an important research and clinical object because of the recent appearance of a new, more pathogenic strain (Ad14P1). In a human epithelial cancer xenograft model, Ad14P1 showed more efficient viral spread and oncolysis than Ad14. Here, we tested the hypothesis that a mutation in the Ad14P1 fiber knob could account for the differences between the two strains. While our X-ray crystallography studies suggested an altered three-dimensional (3D) structure of the Ad14P1 fiber knob in the F-G loop region, this did not significantly change the fiber knob affinity to DSG2 or the intracellular signaling and DSG2 shedding in epithelial cancer cells. IMPORTANCE A number of widely distributed adenoviruses use the epithelial junction protein DSG2 as a receptor for infection and lateral spread. Interaction with DSG2 allows the virus not only to enter cells but also to open epithelial junctions which form a physical barrier to virus spread. Our study elucidates the mechanism beyond virus-triggered junction opening with a focus on adenovirus serotype 3. Ad3 binds to DSG2 with its fiber knob domain and triggers intracellular signaling that culminates in the cleavage of the extracellular domain of DSG2, thereby disrupting DSG2 homodimers between epithelial cells. We confirmed this pathway with a second DSG2-interacting serotype, Ad14, and its recently emerged strain Ad14P1. These new insights in basic adenovirus biology can be employed to develop novel drugs to treat adenovirus infection as well as be used as tools for gene delivery into epithelial tissues or epithelial tumors.
Collapse
|