1
|
Isaguliants M, Zhitkevich A, Petkov S, Gorodnicheva T, Mezale D, Fridrihsone I, Kuzmenko Y, Kostyushev D, Kostyusheva A, Gordeychuk I, Bayurova E. Enzymatic activity of HIV-1 protease defines migration of tumor cells in vitro and enhances their metastatic activity in vivo. Biochimie 2025; 228:32-43. [PMID: 39128490 DOI: 10.1016/j.biochi.2024.08.009] [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: 05/08/2024] [Revised: 07/09/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Overexpression of aspartic proteases, as cathepsin D, is an independent marker of poor prognosis in breast cancer, correlated with the incidence of clinical metastasis. We aimed to find if HIV-1 aspartic protease (PR) can play a similar role. Murine adenocarcinoma 4T1luc2 cells were transduced with lentivirus encoding inactivated drug-resistant PR, generating subclones PR20.1 and PR20.2. Subclones were assessed for production of reactive oxygen species (ROS), expression of epithelial-mesenchymal transition (EMT) factors, and in vitro migratory activity in the presence or absence of antioxidant N-acetyl cysteine and protease inhibitors. Tumorigenic activity was evaluated by implanting cells into BALB/c mice and following tumor growth by calipering and bioluminescence imaging in vivo, and metastases, by organ imaging ex vivo. Both subclones expressed PR mRNA, and PR20.2, also the protein detected by Western blotting. PR did not induce production of ROS, and had no direct effect on cell migration rate, however, treatment with inhibitors of drug-resistant PR suppressed the migratory activity of both subclones. Furthermore, expression of N-cadherin and Vimentin in PR20.2 cells and their migration were enhanced by antioxidant treatment. Sensitivity of in vitro migration to protease inhibitors and to antioxidant, known to restore PR activity, related the effects to the enzymatic activity of PR. In vivo, PR20.2 cells demonstrated higher tumorigenic and metastatic activity than PR20.1 or parental cells. Thus, HIV-1 protease expressed in breast cancer cells determines their migration in vitro and metastatic activity in vivo. This effect may aggravate clinical course of cancers in people living with HIV-1.
Collapse
Affiliation(s)
- M Isaguliants
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - A Zhitkevich
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819, Moscow, Russia.
| | - S Petkov
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - T Gorodnicheva
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia.
| | - D Mezale
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - I Fridrihsone
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - Y Kuzmenko
- Engelhardt Institute of Molecular Biology, Academy of Sciences of the Russian Federation, 119991, Moscow, Russia.
| | - D Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, 119991, Moscow, Russia.
| | - A Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, 119991, Moscow, Russia.
| | - I Gordeychuk
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden; Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819, Moscow, Russia.
| | - E Bayurova
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden; Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819, Moscow, Russia.
| |
Collapse
|
2
|
Kumar G, Saroha B, Kumar S, Kumari B, Arya P, Raghav N, Ghosh S, Nassare VD. 1,2,3‐Triazole‐Based New Aurones as Anticancer Agents with the Capability to Target Extracellular Digestive Enzymes. ChemistrySelect 2024; 9. [DOI: 10.1002/slct.202403885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/07/2024] [Indexed: 01/04/2025]
Abstract
AbstractThis study involves the synthesis of a series of dimethyl substituted novel aurones, featuring 1,2,3‐triazole as an integral structure. All the newly synthesized compounds were thoroughly characterized using various spectroscopic tools and also subjected to computational analysis utilizing the DFT/B3LYP methodology, which involved the determination of frontier molecular orbital energy values and the computation of various quantum chemical parameters. Further their impact on cell viability and cytotoxic activity on the adenocarcinoma gastric cell line (AGS) was investigated using cell‐based MTT assay. Compounds 6d, 6o and 6p displayed significant cytotoxic activity, reducing cell viability to a greater extent with IC50 values of 9.74, 20.09, and 5.92 µM, respectively and even better than the standard chemotherapeutic drug leucovorin (IC50 = 30.8 µM). In addition, all the compounds were also screened for their extracellular enzymatic assay and through in vitro results compound 6n emerged as the efficient inhibitor of amylase (% inhibition = 51.92) and trypsin (% inhibition = 68.36), whereas an activation is observed for lipase (% activation = 269.48). In silico molecular docking was also conducted to assess the interactions between proteins and ligands, revealing the binding patterns of the synthesized compounds and the standard drug with receptor proteins.
Collapse
Affiliation(s)
- Gourav Kumar
- Department of Chemistry Kurukshetra University Kurukshetra Haryana 136119 India
- Biomedical Engineering Department Oregon Health & Science University Portland Oregon 97201 USA
| | - Bhavna Saroha
- Department of Chemistry Kurukshetra University Kurukshetra Haryana 136119 India
- Department of Chemistry Deenbandhu Chhotu Ram University of Science and Technology Murthal Sonipat 131039 India
| | - Suresh Kumar
- Department of Chemistry Kurukshetra University Kurukshetra Haryana 136119 India
| | - Bavita Kumari
- Department of Chemistry Kurukshetra University Kurukshetra Haryana 136119 India
- Department of Chemistry Oregon State University Corvallis Oregon 97331 USA
| | - Priyanka Arya
- Department of Chemistry Kurukshetra University Kurukshetra Haryana 136119 India
| | - Neera Raghav
- Department of Chemistry Kurukshetra University Kurukshetra Haryana 136119 India
| | - Sushmita Ghosh
- Department of Pathology and Cancer Screening Chittaranjan National Cancer Institute 37, S.P. Mukherjee Road Kolkata 700026 India
| | - Vilas D. Nassare
- Department of Pathology and Cancer Screening Chittaranjan National Cancer Institute 37, S.P. Mukherjee Road Kolkata 700026 India
| |
Collapse
|
3
|
Korpidou M, Becker J, Tarvirdipour S, Dinu IA, Becer CR, Palivan CG. Glycooligomer-Functionalized Catalytic Nanocompartments Co-Loaded with Enzymes Support Parallel Reactions and Promote Cell Internalization. Biomacromolecules 2024; 25:4492-4509. [PMID: 38910355 PMCID: PMC11238334 DOI: 10.1021/acs.biomac.4c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024]
Abstract
A major shortcoming associated with the application of enzymes in drug synergism originates from the lack of site-specific, multifunctional nanomedicine. This study introduces catalytic nanocompartments (CNCs) made of a mixture of PDMS-b-PMOXA diblock copolymers, decorated with glycooligomer tethers comprising eight mannose-containing repeating units and coencapsulating two enzymes, providing multifunctionality by their in situ parallel reactions. Beta-glucuronidase (GUS) serves for local reactivation of the drug hymecromone, while glucose oxidase (GOx) induces cell starvation through glucose depletion and generation of the cytotoxic H2O2. The insertion of the pore-forming peptide, melittin, facilitates diffusion of substrates and products through the membranes. Increased cell-specific internalization of the CNCs results in a substantial decrease in HepG2 cell viability after 24 h, attributed to simultaneous production of hymecromone and H2O2. Such parallel enzymatic reactions taking place in nanocompartments pave the way to achieve efficient combinatorial cancer therapy by enabling localized drug production along with reactive oxygen species (ROS) elevation.
Collapse
Affiliation(s)
- Maria Korpidou
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
| | - Jonas Becker
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Shabnam Tarvirdipour
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
| | - Ionel Adrian Dinu
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
| | - C. Remzi Becer
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Cornelia G. Palivan
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
- NCCR
Molecular Systems Engineering, Mattenstrasse 22, Basel 4002, Switzerland
| |
Collapse
|
4
|
Paulus J, Sewald N. Small molecule- and peptide-drug conjugates addressing integrins: A story of targeted cancer treatment. J Pept Sci 2024; 30:e3561. [PMID: 38382900 DOI: 10.1002/psc.3561] [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: 09/20/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 02/23/2024]
Abstract
Targeted cancer treatment should avoid side effects and damage to healthy cells commonly encountered during traditional chemotherapy. By combining small molecule or peptidic ligands as homing devices with cytotoxic drugs connected by a cleavable or non-cleavable linker in peptide-drug conjugates (PDCs) or small molecule-drug conjugates (SMDCs), cancer cells and tumours can be selectively targeted. The development of highly affine, selective peptides and small molecules in recent years has allowed PDCs and SMDCs to increasingly compete with antibody-drug conjugates (ADCs). Integrins represent an excellent target for conjugates because they are overexpressed by most cancer cells and because of the broad knowledge about native binding partners as well as the multitude of small-molecule and peptidic ligands that have been developed over the last 30 years. In particular, integrin αVβ3 has been addressed using a variety of different PDCs and SMDCs over the last two decades, following various strategies. This review summarises and describes integrin-addressing PDCs and SMDCs while highlighting points of great interest.
Collapse
Affiliation(s)
- Jannik Paulus
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Bielefeld, Germany
| |
Collapse
|
5
|
Mitin D, Bullinger F, Dobrynin S, Engelmann J, Scheffler K, Kolokolov M, Krumkacheva O, Buckenmaier K, Kirilyuk I, Chubarov A. Contrast Agents Based on Human Serum Albumin and Nitroxides for 1H-MRI and Overhauser-Enhanced MRI. Int J Mol Sci 2024; 25:4041. [PMID: 38612851 PMCID: PMC11012161 DOI: 10.3390/ijms25074041] [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: 02/27/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
In cancer diagnostics, magnetic resonance imaging (MRI) uses contrast agents to enhance the distinction between the target tissue and background. Several promising approaches have been developed to increase MRI sensitivity, one of which is Overhauser dynamic nuclear polarization (ODNP)-enhanced MRI (OMRI). In this study, a macromolecular construct based on human serum albumin and nitroxyl radicals (HSA-NIT) was developed using a new synthesis method that significantly increased the modification to 21 nitroxide residues per protein. This was confirmed by electron paramagnetic resonance (EPR) spectroscopy and matrix-assisted laser desorption/ionization time-of-flight (MALDI ToF) mass spectrometry. Gel electrophoresis and circular dichroism showed no significant changes in the structure of HSA-NITs, and no oligomers were formed during modification. The cytotoxicity of HSA-NITs was comparable to that of native albumin. HSA-NITs were evaluated as potential "metal-free" organic radical relaxation-based contrast agents for 1H-MRI and as hyperpolarizing contrast agents for OMRI. Relaxivities (longitudinal and transversal relaxation rates r1 and r2) for HSA-NITs were measured at different magnetic field strengths (1.88, 3, 7, and 14 T). Phantoms were used to demonstrate the potential use of HSA-NIT as a T1- and T2-weighted relaxation-based contrast agent at 3 T and 14 T. The efficacy of 1H Overhauser dynamic nuclear polarization (ODNP) in liquids at an ultralow magnetic field (ULF, B0 = 92 ± 0.8 μT) was investigated for HSA-NIT conjugates. The HSA-NITs themselves did not show ODNP enhancement; however, under the proteolysis conditions simulating cancer tissue, HSA-NIT conjugates were cleaved into lower-molecular-weight (MW) protein fragments that activate ODNP capabilities, resulting in a maximum achievable enhancement |Emax| of 40-50 and a radiofrequency power required to achieve half of Emax, P1/2, of 21-27 W. The HSA-NIT with a higher degree of modification released increased the number of spin probes upon biodegradation, which significantly enhanced the Overhauser effect. Thus, HSA-NITs may represent a new class of MRI relaxation-based contrast agents as well as novel cleavable conjugates for use as hyperpolarizing contrast agents (HCAs) in OMRI.
Collapse
Affiliation(s)
- Dmitry Mitin
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia;
| | - Friedemann Bullinger
- High-Field Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany; (F.B.); (J.E.); (K.S.); (K.B.)
| | - Sergey Dobrynin
- N.N. Vorozhtsov Institute of Organic Chemistry SB RAS, 630090 Novosibirsk, Russia;
| | - Jörn Engelmann
- High-Field Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany; (F.B.); (J.E.); (K.S.); (K.B.)
| | - Klaus Scheffler
- High-Field Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany; (F.B.); (J.E.); (K.S.); (K.B.)
- Department of Biomedical Magnetic Resonance, Eberhard-Karls University, 72076 Tuebingen, Germany
| | - Mikhail Kolokolov
- International Tomography Center SB RAS, 630090 Novosibirsk, Russia; (M.K.); (O.K.)
| | - Olesya Krumkacheva
- International Tomography Center SB RAS, 630090 Novosibirsk, Russia; (M.K.); (O.K.)
| | - Kai Buckenmaier
- High-Field Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany; (F.B.); (J.E.); (K.S.); (K.B.)
| | - Igor Kirilyuk
- N.N. Vorozhtsov Institute of Organic Chemistry SB RAS, 630090 Novosibirsk, Russia;
| | - Alexey Chubarov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia;
| |
Collapse
|
6
|
Pang L, Guo S, Khan F, Dunterman M, Ali H, Liu Y, Huang Y, Chen P. Hypoxia-driven protease legumain promotes immunosuppression in glioblastoma. Cell Rep Med 2023; 4:101238. [PMID: 37858339 PMCID: PMC10694605 DOI: 10.1016/j.xcrm.2023.101238] [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: 05/17/2023] [Revised: 08/27/2023] [Accepted: 09/20/2023] [Indexed: 10/21/2023]
Abstract
Glioblastoma (GBM) is a hypoxic and "immune-cold" tumor containing rich stromal signaling molecules and cell populations, such as proteases and immunosuppressive tumor-associated macrophages (TAMs). Here, we seek to profile and characterize the potential proteases that may contribute to GBM immunosuppression. Legumain (LGMN) emerges as the key protease that is highly enriched in TAMs and transcriptionally upregulated by hypoxia-inducible factor 1-alpha (HIF1α). Functionally, the increased LGMN promotes TAM immunosuppressive polarization via activating the GSK-3β-STAT3 signaling pathway. Inhibition of macrophage HIF1α and LGMN reduces TAM immunosuppressive polarization, impairs tumor progression, enhances CD8+ T cell-mediated anti-tumor immunity, and synergizes with anti-PD1 therapy in GBM mouse models. Thus, LGMN is a key molecular switch connecting two GBM hallmarks of hypoxia and immunosuppression, providing an actionable therapeutic intervention for this deadly disease.
Collapse
Affiliation(s)
- Lizhi Pang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Songlin Guo
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Fatima Khan
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Madeline Dunterman
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Heba Ali
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yang Liu
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yuyun Huang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Peiwen Chen
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
7
|
Fejza A, Carobolante G, Poletto E, Camicia L, Schinello G, Di Siena E, Ricci G, Mongiat M, Andreuzzi E. The entanglement of extracellular matrix molecules and immune checkpoint inhibitors in cancer: a systematic review of the literature. Front Immunol 2023; 14:1270981. [PMID: 37854588 PMCID: PMC10579931 DOI: 10.3389/fimmu.2023.1270981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Introduction Immune-checkpoint inhibitors (ICIs) have emerged as a core pillar of cancer therapy as single agents or in combination regimens both in adults and children. Unfortunately, ICIs provide a long-lasting therapeutic effect in only one third of the patients. Thus, the search for predictive biomarkers of responsiveness to ICIs remains an urgent clinical need. The efficacy of ICIs treatments is strongly affected not only by the specific characteristics of cancer cells and the levels of immune checkpoint ligands, but also by other components of the tumor microenvironment, among which the extracellular matrix (ECM) is emerging as key player. With the aim to comprehensively describe the relation between ECM and ICIs' efficacy in cancer patients, the present review systematically evaluated the current literature regarding ECM remodeling in association with immunotherapeutic approaches. Methods This review followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines and was registered at the International Prospective Register of Systematic Reviews (PROSPERO, CRD42022351180). PubMed, Web of Science, and Scopus databases were comprehensively searched from inception to January 2023. Titles, abstracts and full text screening was performed to exclude non eligible articles. The risk of bias was assessed using the QUADAS-2 tool. Results After employing relevant MeSH and key terms, we identified a total of 5070 studies. Among them, 2540 duplicates, 1521 reviews or commentaries were found and excluded. Following title and abstract screening, the full text was analyzed, and 47 studies meeting the eligibility criteria were retained. The studies included in this systematic review comprehensively recapitulate the latest observations associating changes of the ECM composition following remodeling with the traits of the tumor immune cell infiltration. The present study provides for the first time a broad view of the tight association between ECM molecules and ICIs efficacy in different tumor types, highlighting the importance of ECM-derived proteolytic products as promising liquid biopsy-based biomarkers to predict the efficacy of ICIs. Conclusion ECM remodeling has an important impact on the immune traits of different tumor types. Increasing evidence pinpoint at ECM-derived molecules as putative biomarkers to identify the patients that would most likely benefit from ICIs treatments. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022351180, identifier CRD42022351180.
Collapse
Affiliation(s)
- Albina Fejza
- Department of Biochemistry, Faculty of Medical Sciences, UBT-Higher Education Institute, Prishtina, Kosovo
| | - Greta Carobolante
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Lucrezia Camicia
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Giorgia Schinello
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Emanuele Di Siena
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Giuseppe Ricci
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Eva Andreuzzi
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
8
|
Francis D, Veeramanickathadathil Hari G, Koonthanmala Subash A, Bhairaddy A, Joy A. The biofilm proteome of Staphylococcus aureus and its implications for therapeutic interventions to biofilm-associated infections. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:327-400. [PMID: 38220430 DOI: 10.1016/bs.apcsb.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Staphylococcus aureus is a major healthcare concern due to its ability to inflict life-threatening infections and evolve antibiotic resistance at an alarming pace. It is frequently associated with hospital-acquired infections, especially device-associated infections. Systemic infections due to S. aureus are difficult to treat and are associated with significant mortality and morbidity. The situation is worsened by the ability of S. aureus to form social associations called biofilms. Biofilms embed a community of cells with the ability to communicate with each other and share resources within a polysaccharide or protein matrix. S. aureus establish biofilms on tissues and conditioned abiotic surfaces. Biofilms are hyper-tolerant to antibiotics and help evade host immune responses. Biofilms exacerbate the severity and recalcitrance of device-associated infections. The development of a biofilm involves various biomolecules, such as polysaccharides, proteins and nucleic acids, contributing to different structural and functional roles. Interconnected signaling pathways and regulatory molecules modulate the expression of these molecules. A comprehensive understanding of the molecular biology of biofilm development would help to devise effective anti-biofilm therapeutics. Although bactericidal agents, antimicrobial peptides, bacteriophages and nano-conjugated anti-biofilm agents have been employed with varying levels of success, there is still a requirement for effective and clinically viable anti-biofilm therapeutics. Proteins that are expressed and utilized during biofilm formation, constituting the biofilm proteome, are a particularly attractive target for anti-biofilm strategies. The proteome can be explored to identify potential anti-biofilm drug targets and utilized for rational drug discovery. With the aim of uncovering the biofilm proteome, this chapter explores the mechanism of biofilm formation and its regulation. Furthermore, it explores the antibiofilm therapeutics targeted against the biofilm proteome.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India.
| | | | | | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| |
Collapse
|
9
|
Hey G, Rao R, Carter A, Reddy A, Valle D, Patel A, Patel D, Lucke-Wold B, Pomeranz Krummel D, Sengupta S. Ligand-Gated Ion Channels: Prognostic and Therapeutic Implications for Gliomas. J Pers Med 2023; 13:jpm13050853. [PMID: 37241023 DOI: 10.3390/jpm13050853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/05/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Gliomas are common primary brain malignancies that remain difficult to treat due to their overall aggressiveness and heterogeneity. Although a variety of therapeutic strategies have been employed for the treatment of gliomas, there is increasing evidence that suggests ligand-gated ion channels (LGICs) can serve as a valuable biomarker and diagnostic tool in the pathogenesis of gliomas. Various LGICs, including P2X, SYT16, and PANX2, have the potential to become altered in the pathogenesis of glioma, which can disrupt the homeostatic activity of neurons, microglia, and astrocytes, further exacerbating the symptoms and progression of glioma. Consequently, LGICs, including purinoceptors, glutamate-gated receptors, and Cys-loop receptors, have been targeted in clinical trials for their potential therapeutic benefit in the diagnosis and treatment of gliomas. In this review, we discuss the role of LGICs in the pathogenesis of glioma, including genetic factors and the effect of altered LGIC activity on the biological functioning of neuronal cells. Additionally, we discuss current and emerging investigations regarding the use of LGICs as a clinical target and potential therapeutic for gliomas.
Collapse
Affiliation(s)
- Grace Hey
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rohan Rao
- College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Ashley Carter
- Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Akshay Reddy
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Daisy Valle
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Anjali Patel
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Drashti Patel
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 23608, USA
| | - Daniel Pomeranz Krummel
- Department of Neurology & Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Soma Sengupta
- Department of Neurology & Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
10
|
Chemistry towards Biology. Int J Mol Sci 2023; 24:ijms24043998. [PMID: 36835407 PMCID: PMC9960482 DOI: 10.3390/ijms24043998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Although it may not seem like it, chemical biology has existed for a long time from today's perspective [...].
Collapse
|
11
|
Dinca AL, Diaconu A, Birla RD, Coculescu BI, Dinca VG, Manole G, Marica C, Tudorache IS, Panaitescu E, Constantinoiu SM, Coculescu EC. Systemic inflammation factors as survival prognosis markers in ovarian neoplasm and the relationship with cancer-associated inflammatory mediators-a review. Int J Immunopathol Pharmacol 2023; 37:3946320231178769. [PMID: 37246293 DOI: 10.1177/03946320231178769] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023] Open
Abstract
At the level of the genital system, ovarian neoplasm is the most frequent cause of morbidity and mortality. In the specialized literature, the coexistence of an inflammatory process is admitted from the early stages of the evolution of this pathology. Starting from the importance of this process, both in determinism and in the evolution of carcinogenesis and summarizing the field of knowledge, for this study we considered two objectives: the first was the presentation of the pathogenic mechanism, through which chronic +ovarian inflammation is involved in the process of carcinogenesis, and the second is the justification of the clinical utility of the three parameters, accepted as biomarkers of systemic inflammation: neutrophil-lymphocyte ratio, platelet lymphocyte ratio, and lymphocyte-monocyte ratio in the assessment of prognosis. The study highlights the acceptance of these hematological parameters, with practical utility, as prognostic biomarkers in ovarian cancer, based on the intrinsic link with cancer-associated inflammatory mediators. Based on the data from the specialized literature, the conclusion is that in ovarian cancer, the inflammatory process induced by the presence of the tumor, induces changes in the types of circulating leukocytes, with immediate effects on the markers of systemic inflammation.
Collapse
Affiliation(s)
| | - Adriana Diaconu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Rodica Daniela Birla
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Bogdan-Ioan Coculescu
- Faculty of Midwifery and Nursing, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Cantacuzino National Medico-Military Institute for Research and Development, Bucharest, Romania
| | | | - Gheorghe Manole
- Romanian Academy of Medical Sciences
- Faculty of General Nursing, Bioterra University, Bucharest, Romania
| | - Cristian Marica
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Eugenia Panaitescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Elena Claudia Coculescu
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
12
|
Two-phase model of compressive stress induced on a surrounding hyperelastic medium by an expanding tumour. J Math Biol 2022; 86:18. [PMID: 36538075 DOI: 10.1007/s00285-022-01851-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/24/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
In vitro experiments in which tumour cells are seeded in a gelatinous medium, or hydrogel, show how mechanical interactions between tumour cells and the tissue in which they are embedded, together with local levels of an externally-supplied, diffusible nutrient (e.g., oxygen), affect the tumour's growth dynamics. In this article, we present a mathematical model that describes these in vitro experiments. We use the model to understand how tumour growth generates mechanical deformations in the hydrogel and how these deformations in turn influence the tumour's growth. The hydrogel is viewed as a nonlinear hyperelastic material and the tumour is modelled as a two-phase mixture, comprising a viscous tumour cell phase and an isotropic, inviscid interstitial fluid phase. Using a combination of numerical and analytical techniques, we show how the tumour's growth dynamics change as the mechanical properties of the hydrogel vary. When the hydrogel is soft, nutrient availability dominates the dynamics: the tumour evolves to a large equilibrium configuration where the proliferation rate of nutrient-rich cells on the tumour boundary balances the death rate of nutrient-starved cells in the central, necrotic core. As the hydrogel stiffness increases, mechanical resistance to growth increases and the tumour's equilibrium size decreases. Indeed, for small tumours embedded in stiff hydrogels, the inhibitory force experienced by the tumour cells may be so large that the tumour is eliminated. Analysis of the model identifies parameter regimes in which the presence of the hydrogel drives tumour elimination.
Collapse
|
13
|
Masood M, Masood MBE, Us Subah N, Shabbir M, Paracha RZ, Rafiq M. Investigating isoform switching in RHBDF2 and its role in neoplastic growth in breast cancer. PeerJ 2022; 10:e14124. [PMID: 36452073 PMCID: PMC9703992 DOI: 10.7717/peerj.14124] [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: 01/21/2022] [Accepted: 09/06/2022] [Indexed: 11/27/2022] Open
Abstract
Background Breast cancer is the second leading cause of cancer-related deaths globally, and its prevalence rates are increasing daily. In the past, studies predicting therapeutic drug targets for cancer therapy focused on the assumption that one gene is responsible for producing one protein. Therefore, there is always an immense need to find promising and novel anti-cancer drug targets. Furthermore, proteases have an integral role in cell proliferation and growth because the proteolysis mechanism is an irreversible process that aids in regulating cellular growth during tumorigenesis. Therefore, an inactive rhomboid protease known as iRhom2 encoded by the gene RHBDF2 can be considered an important target for cancer treatment. Speculatively, previous studies on gene expression analysis of RHBDF2 showed heterogenous behaviour during tumorigenesis. Consistent with this, several studies have reported the antagonistic role of iRhom2 in tumorigenesis, i.e., either they are involved in negative regulation of EGFR ligands via the ERAD pathway or positively regulate EGFR ligands via the EGFR signalling pathway. Additionally, different opinions suggest iRhom2 mediated cleavage of EGFR ligands takes place TACE dependently or TACE independently. However, reconciling these seemingly opposing roles is still unclear and might be attributed to more than one transcript isoform of iRhom2. Methods To observe the differences at isoform resolution, the current strategy identified isoform switching in RHBDF2 via differential transcript usage using RNA-seq data during breast cancer initiation and progression. Furthermore, interacting partners were found via correlation and enriched to explain their antagonistic role. Results Isoform switching was observed at DCIS, grade 2 and grade 3, from canonical to the cub isoform. Neither EGFR nor ERAD was found enriched. However, pathways leading to TACE-dependent EGFR signalling pathways were more observant, specifically MAPK signalling pathways, GPCR signalling pathways, and toll-like receptor pathways. Nevertheless, it was noteworthy that during CTCs, the cub isoform switches back to the canonical isoform, and the proteasomal degradation pathway and cytoplasmic ribosomal protein pathways were significantly enriched. Therefore, it could be inferred that cub isoform functions during cancer initiation in EGFR signalling. In contrast, during metastasis, where invasion is the primary task, the isoform switches back to the canonical isoform.
Collapse
Affiliation(s)
- Mehar Masood
- School of Interdisciplinary Engineering and Sciences, National University of Sciences and Technology, Islamabad, Pakistan,Faculty of Rehabilitation & Allied Health Sciences, Riphah International University, Islamabad, Pakistan
| | - Madahiah Bint E Masood
- School of Interdisciplinary Engineering and Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Noor Us Subah
- School of Interdisciplinary Engineering and Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Maria Shabbir
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Rehan Zafar Paracha
- School of Interdisciplinary Engineering and Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Mehak Rafiq
- School of Interdisciplinary Engineering and Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| |
Collapse
|
14
|
Ali AA, Al-Othman A, Al-Sayah MH. Multifunctional stimuli-responsive hybrid nanogels for cancer therapy: Current status and challenges. J Control Release 2022; 351:476-503. [PMID: 36170926 DOI: 10.1016/j.jconrel.2022.09.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/18/2022]
Abstract
With cancer research shifting focus to achieving multifunctionality in cancer treatment strategies, hybrid nanogels are making a rapid rise to the spotlight as novel, multifunctional, stimuli-responsive, and biocompatible cancer therapeutic strategies. They can possess cancer cell-specific cytotoxic effects themselves, carry drugs or enzymes that can produce cytotoxic effects, improve imaging modalities, and target tumor cells over normal cells. Hybrid nanogels bring together a wide range of desirable properties for cancer treatment such as stimuli-responsiveness, efficient loading and protection of molecules such as drugs or enzymes, and effective crossing of cellular barriers among other properties. Despite their promising abilities, hybrid nanogels are still far from being used in the clinic, and their available data remains relatively limited. However, many studies can be done to facilitate this clinical transition. This review is critically summarizing and analyzing the recent information and progress on the use of hybrid nanogels particularly inorganic nanoparticle-based and organic nanoparticle-based hybrid nanogels in the field of oncology and future directions to aid in transferring those results to the clinic. This work concludes that the future of hybrid nanogels is greatly impacted by therapeutic and non-therapeutic factors. Therapeutic factors include the lack of hemocompatibility studies, acute and chronic toxicological studies, and information on agglomeration capability and extent, tumor heterogeneity, interaction with proteins in physiological fluids, endocytosis-exocytosis, and toxicity of the nanogels' breakdown products. Non-therapeutic factors include the lack of clear regulatory guidelines and standardized assays, limitations of animal models, and difficulties associated with good manufacture practices (GMP).
Collapse
Affiliation(s)
- Amaal Abdulraqeb Ali
- Biomedical Engineering Graduate Program, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates
| | - Amani Al-Othman
- Department of Chemical Engineering, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates.
| | - Mohammad H Al-Sayah
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates
| |
Collapse
|
15
|
Inhibition of Colon Cancer Recurrence via Exogenous TRAIL Delivery Using Gel-like Coacervate Microdroplets. Gels 2022; 8:gels8070427. [PMID: 35877512 PMCID: PMC9319433 DOI: 10.3390/gels8070427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
Colon cancer (CC) belongs to the three major malignancies with a high recurrence rate. Therefore, a novel drug delivery system that can prevent CC recurrence while minimizing side effects is needed. Tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL) has recently been spotlighted as a protein drug that can induce apoptosis of cancer cells specifically. However, its short in vivo half-life is still a challenge to overcome. Hence, in this study, a gel-like mPEGylated coacervate (mPEG-Coa) delivery platform was developed through electrostatic interaction of mPEG-poly(ethylene arginylaspartate diglyceride) (mPEG-PEAD) and heparin for effective protection of cargo TRAIL, subsequently preserving its bioactivity. mPEG-Coa could protect cargo TRAIL against protease. Sustained release was observed for a long-term (14 days). In addition, recurrence of HCT-116 cells was suppressed when cells were treated with TRAIL-loaded mPEG-Coa for 7 days through long-term continuous supply of active TRAIL, whereas re-proliferation occurred in the bolus TRAIL-treated group. Taken together, these results suggest that our gel-like mPEG-Coa could be utilized as a functional delivery platform to suppress CC recurrence by exogenously supplying TRAIL for a long time with a single administration.
Collapse
|
16
|
Ben-Nissan G, Katzir N, Füzesi-Levi MG, Sharon M. Biology of the Extracellular Proteasome. Biomolecules 2022; 12:619. [PMID: 35625547 PMCID: PMC9139032 DOI: 10.3390/biom12050619] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022] Open
Abstract
Proteasomes are traditionally considered intracellular complexes that play a critical role in maintaining proteostasis by degrading short-lived regulatory proteins and removing damaged proteins. Remarkably, in addition to these well-studied intracellular roles, accumulating data indicate that proteasomes are also present in extracellular body fluids. Not much is known about the origin, biological role, mode(s) of regulation or mechanisms of extracellular transport of these complexes. Nevertheless, emerging evidence indicates that the presence of proteasomes in the extracellular milieu is not a random phenomenon, but rather a regulated, coordinated physiological process. In this review, we provide an overview of the current understanding of extracellular proteasomes. To this end, we examine 143 proteomic datasets, leading us to the realization that 20S proteasome subunits are present in at least 25 different body fluids. Our analysis also indicates that while 19S subunits exist in some of those fluids, the dominant proteasome activator in these compartments is the PA28α/β complex. We also elaborate on the positive correlations that have been identified in plasma and extracellular vesicles, between 20S proteasome and activity levels to disease severity and treatment efficacy, suggesting the involvement of this understudied complex in pathophysiology. In addition, we address the considerations and practical experimental methods that should be taken when investigating extracellular proteasomes. Overall, we hope this review will stimulate new opportunities for investigation and thoughtful discussions on this exciting topic that will contribute to the maturation of the field.
Collapse
Affiliation(s)
| | | | | | - Michal Sharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel; (G.B.-N.); (N.K.); (M.G.F.-L.)
| |
Collapse
|
17
|
Casazza A, Van Helleputte L, Van Renterghem B, Pokreisz P, De Geest N, De Petrini M, Janssens T, Pellens M, Diricx M, Riera-Domingo C, Wozniak A, Mazzone M, Schöffski P, Defert O, Reyns G, Kindt N. PhAc-ALGP-Dox, a Novel Anticancer Prodrug with Targeted Activation and Improved Therapeutic Index. Mol Cancer Ther 2022; 21:568-581. [PMID: 35149549 PMCID: PMC9377749 DOI: 10.1158/1535-7163.mct-21-0518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/13/2021] [Accepted: 02/08/2022] [Indexed: 01/07/2023]
Abstract
Clinical use of doxorubicin (Dox) is limited by cumulative myelo- and cardiotoxicity. This research focuses on the detailed characterization of PhAc-ALGP-Dox, a targeted tetrapeptide prodrug with a unique dual-step activation mechanism, designed to circumvent Dox-related toxicities and is ready for upcoming clinical investigation. Coupling Dox to a phosphonoacetyl (PhAc)-capped tetrapeptide forms the cell-impermeable, inactive compound, PhAc-ALGP-Dox. After extracellular cleavage by tumor-enriched thimet oligopeptidase-1 (THOP1), a cell-permeable but still biologically inactive dipeptide-conjugate is formed (GP-Dox), which is further processed intracellularly to Dox by fibroblast activation protein-alpha (FAPα) and/or dipeptidyl peptidase-4 (DPP4). In vitro, PhAc-ALGP-Dox is effective in various 2D- and 3D-cancer models, while showing improved safety toward normal epithelium, hematopoietic progenitors, and cardiomyocytes. In vivo, these results translate into a 10-fold higher tolerability and 5-fold greater retention of Dox in the tumor microenvironment compared with the parental drug. PhAc-ALGP-Dox demonstrates 63% to 96% tumor growth inhibition in preclinical models, an 8-fold improvement in efficacy in patient-derived xenograft (PDX) models, and reduced metastatic burden in a murine model of experimental lung metastasis, improving survival by 30%. The current findings highlight the potential clinical benefit of PhAc-ALGP-Dox, a targeted drug-conjugate with broad applicability, favorable tissue biodistribution, significantly improved tolerability, and tumor growth inhibition at primary and metastatic sites in numerous solid tumor models.
Collapse
Affiliation(s)
- Andrea Casazza
- CoBioRes NV, Campus Gasthuisberg University of Leuven, Leuven, Belgium
| | | | - Britt Van Renterghem
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Peter Pokreisz
- CoBioRes NV, Campus Gasthuisberg University of Leuven, Leuven, Belgium
| | - Natalie De Geest
- CoBioRes NV, Campus Gasthuisberg University of Leuven, Leuven, Belgium
| | - Marzia De Petrini
- CoBioRes NV, Campus Gasthuisberg University of Leuven, Leuven, Belgium
| | - Tom Janssens
- CoBioRes NV, Campus Gasthuisberg University of Leuven, Leuven, Belgium
| | - Marijke Pellens
- CoBioRes NV, Campus Gasthuisberg University of Leuven, Leuven, Belgium
| | - Marjan Diricx
- CoBioRes NV, Campus Gasthuisberg University of Leuven, Leuven, Belgium
| | - Carla Riera-Domingo
- Laboratory of Tumor Inflammation and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium.,Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Agnieszka Wozniak
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium.,Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Patrick Schöffski
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven, Belgium.,Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Olivier Defert
- CoBioRes NV, Campus Gasthuisberg University of Leuven, Leuven, Belgium
| | - Geert Reyns
- CoBioRes NV, Campus Gasthuisberg University of Leuven, Leuven, Belgium
| | - Nele Kindt
- CoBioRes NV, Campus Gasthuisberg University of Leuven, Leuven, Belgium.,Corresponding Author: Nele Kindt, CoBioRes NV, Campus Gasthuisberg, CDG, bus 913 Herestraat 49, Leuven, Flanders B-3000, Belgium. E-mail:
| |
Collapse
|