1
|
Mizuno Y, Suebboonprathueng T, Onoe S, Akizawa H, Nishijima KI, Takahashi K, Kuge Y. Design of a Tetravalent RGD Peptide Capable of Simultaneous Binding with Multiple Integrin αvβ3 for Targeted Radionuclide Therapy. J Med Chem 2025; 68:6518-6533. [PMID: 40083181 DOI: 10.1021/acs.jmedchem.4c03007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
For targeted radionuclide therapy, radioligands that exhibit high and persistent tumor uptake are indispensable. We previously synthesized a 99mTc-labeled hexavalent RGD peptide (99mTc-(RGD)6) as a tumor imaging agent targeting integrin αvβ3. 99mTc-(RGD)6 showed high in vivo tumor uptake with long retention due to simultaneous binding to multiple integrin αvβ3 receptors. The purpose of this study was to apply this finding to the design of a multivalent RGD peptide labeled with 211At, a promising α-emitting radionuclide for radionuclide therapy. As a candidate compound, a tetravalent RGD peptide (H2N-(RGD)4) was synthesized and radiolabeled with 125I, a homologous element of At, for basic studies. As expected, 125I-(RGD)4 retained the capability of simultaneous binding and showed comparable in vivo tumor uptake to 99mTc-(RGD)6. Finally, 211At-(RGD)4 was synthesized with >95% radiochemical purity and exhibited an almost identical biodistribution pattern to 125I-(RGD)4. These results indicate that 211At-(RGD)4 might be a potential radioligand for integrin αvβ3-targeted radionuclide therapy.
Collapse
Affiliation(s)
- Yuki Mizuno
- Central Institute of Isotope Science, Hokkaido University, Sapporo 060-0815, Japan
- Global Center for Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Japan
| | | | - Satoru Onoe
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, Machida 194-8543, Japan
| | - Hiromichi Akizawa
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, Machida 194-8543, Japan
| | - Ken-Ichi Nishijima
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Sapporo 060-0815, Japan
- Global Center for Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Japan
| |
Collapse
|
2
|
Jia F, Peng Y, Li X, Yang S, Xie Y, Han Y, Huang M, Liu T, Zou W, Chen L, Liang Z. Matrix metallopeptidase 2-responsive curcumin-loaded nanoparticles-induced signal transducer and activator of transcription 3 inhibition suppresses glioblastoma multiforme growth via enhancing nuclear factor erythroid 2-related factor 2 activity. Int J Biol Macromol 2025; 307:141998. [PMID: 40081725 DOI: 10.1016/j.ijbiomac.2025.141998] [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: 12/03/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
This study investigated the inhibitory effects of matrix metallopeptidase 2 (MMP2)-responsive curcumin-loaded nanoparticles on glioblastoma multiforme (GBM), and elucidated their underlying mechanisms. The methods employed included the Cell Counting Kit-8 viability assay, colony formation assay, flow cytometry for apoptosis analysis, wound healing migration assay, quantitative real-time polymerase chain reaction, western blotting for gene expression profiling, mitochondrial function assessment, and in vivo antitumor efficacy evaluation. Curcumin significantly reduced the viability, proliferation, and migratory capacity of murine glioma cells (GL261). It also induced apoptosis, disrupted mitochondrial function, and increased reactive oxygen species levels. Notably, curcumin upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) expression while inhibiting signal transducer and activator of transcription 3 (STAT3) activation. The synthesized MMP2-responsive curcumin nanoparticles (Cur-NPs) effectively suppressed tumor growth and prolonged survival in a GBM mouse model. These data suggest that curcumin inhibits STAT3 activity via an Nrf2-dependent mechanism. This study advances our understanding of the mechanism of action of curcumin and suggests potential avenues for the development of targeted therapies for GBM.
Collapse
Affiliation(s)
- Fujie Jia
- Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519020, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Yingpeng Peng
- The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519020, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Xuanzi Li
- The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519020, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Shuai Yang
- The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519020, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Yuping Xie
- Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519020, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Yuanyuan Han
- Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519020, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Mingsheng Huang
- Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519020, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Tingyao Liu
- Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519020, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Wei Zou
- Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519020, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China.
| | - Lei Chen
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519020, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China.
| | - Zibin Liang
- Department of Thoracic Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519020, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China.
| |
Collapse
|
3
|
Jastrząb P, Car H, Wielgat P. Cell membrane sialome machinery and regulation of receptor tyrosine kinases in gliomas: The functional relevance and therapeutic perspectives. Biomed Pharmacother 2025; 184:117921. [PMID: 39986236 DOI: 10.1016/j.biopha.2025.117921] [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: 12/01/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025] Open
Abstract
Gliomas are the most common primary brain tumors characterized by high aggressive potential, poor therapeutic response, and significantly reduced overall patient survival. Despite significant progress in the diagnosis and therapy of cancer, gliomas remain a clinical challenge due to the high molecular and cellular heterogeneity, which provides for multiple mechanisms of chemoresistance and adaptive plasticity. A better understanding of cellular regulatory mechanisms of intracellular signal transduction enables the development of targeted drug therapies and clinical application. The increasing evidence confirms the role of sialoglycans in the processing of cell membrane receptors via altered dimerization, activation, and autophosphorylation, which results in changes in cellular signaling and promotes cancer progression. Hence, the modified sialylation patterns, as a hallmark of cancer, have been described as modulators of chemotherapy effectiveness and drug resistance. The receptor tyrosine kinases (RTKs)-mediated signaling in glial tumors control cell growth, survival, migration, and angiogenesis. Here, we focus on the engagement of the sialome machinery in RTKs processing in gliomas and its importance as a suitable therapeutic target. The analysis of the sialylation pattern and its impact on the activity of growth factor receptors provides valuable insights into our understanding of the molecular and cellular complexity of glial tumors. This highlights the novel treatment approaches that could improve prognosis and patients' overall survival.
Collapse
Affiliation(s)
- Patrycja Jastrząb
- Department of Clinical Pharmacology, Medical University of Bialystok, ul. Waszyngtona 15A, Bialystok 15-274, Poland
| | - Halina Car
- Department of Clinical Pharmacology, Medical University of Bialystok, ul. Waszyngtona 15A, Bialystok 15-274, Poland; Department of Experimental Pharmacology, Medical University of Bialystok, ul. Szpitalna 37, Bialystok 15-295, Poland
| | - Przemyslaw Wielgat
- Department of Clinical Pharmacology, Medical University of Bialystok, ul. Waszyngtona 15A, Bialystok 15-274, Poland.
| |
Collapse
|
4
|
Zhuo P, Li Q, Yang B, Li N, Luo Z, Zhang F. Interaction of integrin α vβ 3 and fibronectin under fluid shear forces: implications for tumor cell adhesion and migration. Front Cell Dev Biol 2025; 13:1512672. [PMID: 40070879 PMCID: PMC11894259 DOI: 10.3389/fcell.2025.1512672] [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: 10/17/2024] [Accepted: 01/24/2025] [Indexed: 03/14/2025] Open
Abstract
The interaction between integrin αvβ3 and fibronectin enables tumor cell adherence to endothelial layers under diverse hydrodynamic blood flow conditions, particularly in low shear stress regions. Understanding the mechanical binding characteristics between integrin αvβ3 and fibronectin under different hydrodynamic environments can provide insights into tumor cell invasion and proliferation. Here, the adhesive behavior of fibronectin-functionalized microspheres on integrin αvβ3-coated substrates under various wall fluid shear forces (0.1-0.7 dyn/cm2) was assessed using a parallel plate flow chamber system. The bond lifetimes of integrin αvβ3-fibronectin initially increased and then decreased, indicating transition from a "catch bond" to "slip bond." Upon perfusion of fibronectin-coated microspheres into flow chambers with high-density integrin αvβ3 coating, the rolling velocity of the microspheres increased with increasing shear force. Additionally, the mean stop time and stop frequency exhibited a force-dependent biphasic pattern, initially increasing and then decreasing with shear force, demonstrating a nuanced response to mechanical forces. Thus, the integrin αvβ3-fibronectin interaction displays a "catch bond" property, influencing cell distribution in varying fluid shear forces by promoting optimal adhesion in specific shear sites. These insights enhance our understanding of tumor cell adhesion and migration in hydrodynamic environments and may aid the design of integrin αvβ3-targeted therapies.
Collapse
Affiliation(s)
- Paimin Zhuo
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Quhuan Li
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Bishan Yang
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Na Li
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Zhiqing Luo
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Fengxia Zhang
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
5
|
Santoro F, Merlino F, Brancaccio D, Camerino I, Belli S, Cimmino A, Grieco P, Colucci-D’Amato L, Stoppelli MP, Franco P, Carotenuto A. Glioblastoma Cell Migration, Invasion and Vasculogenic Mimicry Downmodulated by Novel uPAcyclin Derivatives. Cells 2025; 14:259. [PMID: 39996732 PMCID: PMC11853379 DOI: 10.3390/cells14040259] [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: 11/27/2024] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Despite extensive efforts to develop new treatments, the prognosis for glioblastoma multiforme (GBM) is extremely unfavorable, urging the identification of new chemotherapeutics. A previous work identified the cyclic decapeptide uPAcyclin as a potent inhibitor of GBM cell migration, matrix invasion and vascular-like structures' formation, acting through binding to αV integrins and not interfering with cell proliferation or survival. These clearcut activities prompted us to design and test novel derivatives on cultured U87-MG and U251 GBM-MG human cells. With the exception of the residues involved in peptide cyclization, residues were Ala-substituted one by one and the single peptides tested for binding affinity for the αV target integrin, the inhibition of migration, invasion and vasculogenic mimicry. The first screening highlighted peptides with a low binding affinity and low inhibitory ability (Ala4,7,9 derivatives) and peptides with affinity and inhibitory capacity higher than uPAcyclin (Ala2,5,6,8 derivatives). The integration of these results with conformational studies led to the design of the di-substituted variant uPAcyclin. Intriguingly, at least ten-fold greater anti-migratory and anti-invasive effects of the [Ala2,Ala5]uPAcyclin variant compared to uPAcyclin were found. The latter variant also exhibited a greater inhibitory potential for vascular-like structures' formation by matrix-seeded GBM cells. These studies shed light on the functional relevance of single amino acid residues in uPAcyclin and lead to the identification of therapeutically interesting new variants as promising candidates for anti-GBM therapies.
Collapse
Affiliation(s)
- Federica Santoro
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (F.S.); (F.M.); (D.B.); (P.G.)
- Centro Interuniversitario di Ricerca sui Peptidi Bioattivi “Carlo Pedone” (CIRPeB), University of Naples Federico II, 80134 Naples, Italy
| | - Francesco Merlino
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (F.S.); (F.M.); (D.B.); (P.G.)
- Centro Interuniversitario di Ricerca sui Peptidi Bioattivi “Carlo Pedone” (CIRPeB), University of Naples Federico II, 80134 Naples, Italy
| | - Diego Brancaccio
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (F.S.); (F.M.); (D.B.); (P.G.)
| | - Iolanda Camerino
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (I.C.); (L.C.-D.)
| | - Stefania Belli
- Institute of Genetics and Biophysics “A. Buzzati Traverso” (IGB-ABT), National Research Council, 80131 Naples, Italy; (S.B.); (A.C.); (M.P.S.); (P.F.)
| | - Amelia Cimmino
- Institute of Genetics and Biophysics “A. Buzzati Traverso” (IGB-ABT), National Research Council, 80131 Naples, Italy; (S.B.); (A.C.); (M.P.S.); (P.F.)
| | - Paolo Grieco
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (F.S.); (F.M.); (D.B.); (P.G.)
- Centro Interuniversitario di Ricerca sui Peptidi Bioattivi “Carlo Pedone” (CIRPeB), University of Naples Federico II, 80134 Naples, Italy
| | - Luca Colucci-D’Amato
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (I.C.); (L.C.-D.)
- InterUniversity Center for Research in Neurosciences (CIRN), 80131 Naples, Italy
| | - Maria Patrizia Stoppelli
- Institute of Genetics and Biophysics “A. Buzzati Traverso” (IGB-ABT), National Research Council, 80131 Naples, Italy; (S.B.); (A.C.); (M.P.S.); (P.F.)
- Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Paola Franco
- Institute of Genetics and Biophysics “A. Buzzati Traverso” (IGB-ABT), National Research Council, 80131 Naples, Italy; (S.B.); (A.C.); (M.P.S.); (P.F.)
| | - Alfonso Carotenuto
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (F.S.); (F.M.); (D.B.); (P.G.)
- Centro Interuniversitario di Ricerca sui Peptidi Bioattivi “Carlo Pedone” (CIRPeB), University of Naples Federico II, 80134 Naples, Italy
| |
Collapse
|
6
|
Weerarathna IN, Kumar P, Luharia A, Mishra G. Engineering with Biomedical Sciences Changing the Horizon of Healthcare-A Review. Bioengineered 2024; 15:2401269. [PMID: 39285709 PMCID: PMC11409512 DOI: 10.1080/21655979.2024.2401269] [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: 11/30/2023] [Revised: 03/20/2024] [Accepted: 07/18/2024] [Indexed: 01/16/2025] Open
Abstract
In the dynamic realm of healthcare, the convergence of engineering and biomedical sciences has emerged as a pivotal frontier. In this review we go into specific areas of innovation, including medical imaging and diagnosis, developments in biomedical sensors, and drug delivery systems. Wearable biosensors, non-wearable biosensors, and biochips, which include gene chips, protein chips, and cell chips, are all included in the scope of the topic that pertains to biomedical sensors. Extensive research is conducted on drug delivery systems, spanning topics such as the integration of computer modeling, the optimization of drug formulations, and the design of delivery devices. Furthermore, the paper investigates intelligent drug delivery methods, which encompass stimuli-responsive systems such as temperature, redox, pH, light, enzyme, and magnetic responsive systems. In addition to that, the review goes into topics such as tissue engineering, regenerative medicine, biomedical robotics, automation, biomechanics, and the utilization of green biomaterials. The purpose of this analysis is to provide insights that will enhance continuing research and development efforts in engineering-driven biomedical breakthroughs, ultimately contributing to the improvement of healthcare. These insights will be provided by addressing difficulties and highlighting future prospects.
Collapse
Affiliation(s)
- Induni N. Weerarathna
- School of Allied Health Sciences, Department of Biomedical Sciences, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| | - Praveen Kumar
- Department of Computer Science and Medical Engineering, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| | - Anurag Luharia
- Department of Radio Physicist and Radio Safety, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| | - Gaurav Mishra
- Department of Radio Diagnosis, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India
| |
Collapse
|
7
|
Franco MS, Raulefs S, Schilling D, Combs SE, Schmid TE. Impact of Radiation on Invasion and Migration of Glioma In Vitro and In Vivo. Cancers (Basel) 2024; 16:3900. [PMID: 39682088 DOI: 10.3390/cancers16233900] [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: 10/18/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Glioblastoma (GBM) constitutes the most common primary brain tumor and it remains incurable despite therapeutic advances. The high infiltration/invasion potential of GBM cells is considered to be one of the reasons for the inevitable recurrence of the disease. Radiotherapy (RT) is part of the standard care for patients with GBM, and its benefits on overall survival are extensively reported. However, numerous preclinical studies show that X-ray irradiation can enhance the motility of GBM cells. In the present review, we bring together state-of-the-art research on the impact of radiation on GBM cell motility. The mechanisms through which irradiation impacts the brain tumor microenvironment and the tumor cells themselves, leading to more aggressive/invasive tumors, are described. Finally, we summarize potential pharmacological strategies to overcome this problem. Clinical data validating the occurrence of these processes are urgently needed as they could be of great value for patient outcomes. With this comprehensive review, we expect to highlight the need for methods which allow for monitoring the post-irradiation invasive behavior of GBM in patients.
Collapse
Affiliation(s)
- Marina Santiago Franco
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Susanne Raulefs
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Daniela Schilling
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Stephanie E Combs
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Thomas E Schmid
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| |
Collapse
|
8
|
Szivos L, Virga J, Mészár Z, Rostás M, Bakó A, Zahuczki G, Hortobágyi T, Klekner Á. Prognostic Role of Invasion-Related Extracellular Matrix Molecules in Diffusely Infiltrating Grade 2 and 3 Astrocytomas. Brain Sci 2024; 14:1157. [PMID: 39595920 PMCID: PMC11592374 DOI: 10.3390/brainsci14111157] [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: 10/18/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Astrocytoma, an IDH-mutant is a common primary brain tumor. Total surgical resection is not feasible due to peritumoral infiltration mediated by extracellular matrix (ECM) molecules. METHODS This study aimed at determining the expression pattern of ECM molecules in different prognostic groups of WHO grade 2 and grade 3 patients and identifying the effect of onco-radiotherapy on tumor cell invasion of grade 3 patients. Gene and protein expression of ECM molecules was determined by qRT-PCR and immunohistochemistry, respectively. RESULTS In the different prognostic groups of grade 2 tumors HMMR, IDH-1, MKI-67, PDGF-A and versican, in grade 3 tumors integrin α-3, and in both groups integrin α-3 and IDH-1 mRNA expression was significantly different. Regarding protein expression, only integrin αV expression changed significantly in the prognostic groups of grade 2 tumors. CONCLUSIONS Based on the invasion spectrum determined by this joint gene and protein expression analysis, there was a sensitivity of 87.5% and a negative predictive value of 88.9% regarding the different prognostic groups of grade 2 astrocytoma. For grade 3 tumors, the applied standard oncotherapeutic modalities apparently lacked significant anti-invasive effects.
Collapse
Affiliation(s)
- László Szivos
- Department of Neurosurgery, University of Debrecen, H-4032 Debrecen, Hungary or (L.S.); (J.V.)
- Department of Neurosurgery, University of Szeged, H-6725 Szeged, Hungary
| | - József Virga
- Department of Neurosurgery, University of Debrecen, H-4032 Debrecen, Hungary or (L.S.); (J.V.)
- Department of Oncology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Zoltán Mészár
- Department of Anatomy, Histology and Embryology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Melinda Rostás
- Department of Biochemistry and Molecular Biology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Andrea Bakó
- Department of Oncology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Gábor Zahuczki
- UD-GenoMed Medical Genomic Technologies Ltd., H-4032 Debrecen, Hungary;
| | - Tibor Hortobágyi
- Department of Neurology, University of Debrecen, H-4032 Debrecen, Hungary;
- Institute of Neuropathology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Álmos Klekner
- Department of Neurosurgery, University of Debrecen, H-4032 Debrecen, Hungary or (L.S.); (J.V.)
| |
Collapse
|
9
|
Gotorbe C, Segui F, Echavidre W, Durivault J, Blanchard T, Vial V, Pagnuzzi-Boncompagni M, Villeneuve R, Amblard R, Garnier N, Ortholan C, Serrano B, Picco V, Pouysségur J, Vucetic M, Montemagno C. Exploiting Integrin-αVβ3 to Enhance Radiotherapy Efficacy in Medulloblastoma via Ferroptosis. Curr Oncol 2024; 31:7390-7402. [PMID: 39590175 PMCID: PMC11592711 DOI: 10.3390/curroncol31110545] [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: 09/20/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Medulloblastoma, a malignant pediatric brain tumor, has a poor prognosis upon relapse, highlighting a critical clinical need. Our previous research linked medulloblastoma cell radioresistance to integrin-αvβ3 expression. β3-depleted (β3_KO) medulloblastoma cells exhibit lipid hydroxyperoxide accumulation after radiotherapy, indicating ferroptosis, a regulated cell death induced by ROS and inhibited by antioxidants such as cysteine, glutathione (GSH), and glutathione peroxidase 4 (GPx4). However, the link between αvβ3 expression, ferroptosis inhibition, and sensitivity to radiotherapy remains unclear. We showed that irradiated β3_KO medulloblastoma cells primarily die by ferroptosis, with β3-subunit expression correlating with radiotherapy sensitivity and anti-ferroptotic protein levels. Our findings suggest that integrin-αvβ3 signaling boosts oxidative stress resilience via mTORC1. Thus, targeting integrin-αvβ3 could enhance radiotherapy efficacy in medulloblastoma by inducing ferroptotic cell death.
Collapse
Affiliation(s)
- Célia Gotorbe
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Fabien Segui
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - William Echavidre
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Jérôme Durivault
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Thays Blanchard
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Valérie Vial
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Marina Pagnuzzi-Boncompagni
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Rémy Villeneuve
- Radiophysics Department, Princess Grace Hospital, 98000 Monaco, Monaco; (R.V.); (R.A.); (N.G.); (B.S.)
| | - Régis Amblard
- Radiophysics Department, Princess Grace Hospital, 98000 Monaco, Monaco; (R.V.); (R.A.); (N.G.); (B.S.)
| | - Nicolas Garnier
- Radiophysics Department, Princess Grace Hospital, 98000 Monaco, Monaco; (R.V.); (R.A.); (N.G.); (B.S.)
| | - Cécile Ortholan
- Radiotherapy Department, Princess Grace Hospital, 98000 Monaco, Monaco;
| | - Benjamin Serrano
- Radiophysics Department, Princess Grace Hospital, 98000 Monaco, Monaco; (R.V.); (R.A.); (N.G.); (B.S.)
| | - Vincent Picco
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Jacques Pouysségur
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
- CNRS, INSERM, Centre A. Lacassagne, Institute for Research on Cancer & Aging (IRCAN), University Côte d’Azur, 06107 Nice, France
| | - Milica Vucetic
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Christopher Montemagno
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| |
Collapse
|
10
|
Granados-Aparici S, Vieco-Martí I, López-Carrasco A, Navarro S, Noguera R. Real-time morphometric analysis of targeted therapy for neuroblastoma cells in monolayer and 3D hydrogels using digital holographic microscopy. iScience 2024; 27:111231. [PMID: 39569369 PMCID: PMC11576390 DOI: 10.1016/j.isci.2024.111231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/26/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
High-risk neuroblastoma (HR-NB) patient treatment is currently insufficient and challenging due to its high clinical, morphological, and genetic heterogeneity as well as the scarcity of available samples for research. We used a gelatin- and silk fibroin-based hydrogel system with cross-linked vitronectin (VN) as an artificial biomimetic three-dimensional (3D) environment to mirror aggressive neuroblastoma (NB) tumors and tested long-term cell response to Cilengitide (CLG). Based on our previous studies and others using the integrin inhibitor CLG as a potential mechanotherapy drug, we show that CLG caused cell detachment in monolayer cultures of MYCN-amplified SK-N-BE (2) and ALK-mutated SH-SY5Y human neuroblastoma cell lines. Cell detachment and aggregation were maintained in hydrogel-free monolayer cells whereas cells embedded in hydrogels presented different responses to treatment, suggesting differential anoikis resistance between the two cell lines. This underscores the advantages of testing therapeutic approaches using real-time imaging of tumor cells in 3D biomimetic models and its contribution to precision medicine.
Collapse
Affiliation(s)
- Sofia Granados-Aparici
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Isaac Vieco-Martí
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Amparo López-Carrasco
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Samuel Navarro
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rosa Noguera
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
11
|
Cunha Silva L, Branco F, Cunha J, Vitorino C, Gomes C, Carrascal MA, Falcão A, Miguel Neves B, Teresa Cruz M. The potential of exosomes as a new therapeutic strategy for glioblastoma. Eur J Pharm Biopharm 2024; 203:114460. [PMID: 39218361 DOI: 10.1016/j.ejpb.2024.114460] [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: 10/03/2023] [Revised: 07/30/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Glioblastoma (GBM) stands for the most common and aggressive type of brain tumour in adults. It is highly invasive, which explains its short rate of survival. Little is known about its risk factors, and current therapy is still ineffective. Hence, efforts are underway to develop novel and effective treatment approaches against this type of cancer. Exosomes are being explored as a promising strategy for conveying and delivering therapeutic cargo to GBM cells. They can fuse with the GBM cell membrane and, consequently, serve as delivery systems in this context. Due to their nanoscale size, exosomes can cross the blood-brain barrier (BBB), which constitutes a significant hurdle to most chemotherapeutic drugs used against GBM. They can subsequently inhibit oncogenes, activate tumour suppressor genes, induce immune responses, and control cell growth. However, despite representing a promising tool for the treatment of GBM, further research and clinical studies regarding exosome biology, engineering, and clinical applications still need to be completed. Here, we sought to review the application of exosomes in the treatment of GBM through an in-depth analysis of the scientific and clinical studies on the entire process, from the isolation and purification of exosomes to their design and transformation into anti-oncogenic drug delivery systems. Surface modification of exosomes to enhance BBB penetration and GBM-cell targeting is also a topic of discussion.
Collapse
Affiliation(s)
- Leonor Cunha Silva
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Francisco Branco
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Joana Cunha
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3004 535, Portugal
| | - Célia Gomes
- Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal; Center for Innovation in Biomedicine and Biotechnology, CIBB, University of Coimbra, Coimbra 3000-504, Portugal
| | - Mylène A Carrascal
- Tecnimede Group, Sintra 2710-089, Portugal; Center for Neuroscience and Cell Biology, CNC, University of Coimbra, Coimbra 3004-504, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research, CIBIT, University of Coimbra, Coimbra 3000-548, Portugal
| | - Bruno Miguel Neves
- Department of Medical Sciences and Institute of Biomedicine, iBiMED, University of Aveiro, Aveiro 3810-193, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal; Center for Neuroscience and Cell Biology, CNC, University of Coimbra, Coimbra 3004-504, Portugal.
| |
Collapse
|
12
|
Mariasoosai C, Bose S, Natesan S. Structural insights into the molecular recognition of integrin αVβ3 by RGD-containing ligands: The role of the specificity-determining loop (SDL). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614545. [PMID: 39386435 PMCID: PMC11463590 DOI: 10.1101/2024.09.23.614545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Integrin αVβ3 is a prominent member of the "RGD-recognizing" integrin family of cell surface receptors. αVβ3 binds to various extracellular matrix (ECM) proteins and oxysterols such as 25-hydroxycholesterol, is implicated in several diseases, including cancer metastasis, lung fibrosis, inflammation, and autoimmune diseases, and is pursued as a valuable therapeutic target. Despite enormous efforts to seek a pure antagonist, to date, no single drug candidate has successfully reached clinics due to associated partial agonism and toxicity issues. Developing effective and safe inhibitors require a thorough understanding of the molecular interactions and structural changes related to the receptor's activation and inhibition mechanisms. This study offers a comprehensive residue-residue contact and network analyses of the ligand-binding β-propeller βI domains (headpiece) based on all available experimental structures of integrin αVβ3 in unliganded, agonist-, antagonist-, and antibody-bound states. The analyses reveal many critical interactions that were not reported before and show that specific orientation and interactions of residues from the specificity-determining loop (SDL) are critical in molecular recognition and regulation. Also, the network analysis reveals that residues from the nearby allosteric site (site II) connect to the primary RGD-binding site via SDL, which likely acts as an interface between the two sites. Our results provide valuable insights into molecular interactions, structural changes, distinct features of the active and inactive headpiece conformations, the role of SDL in ligand recognition, and SDL-mediated allostery. Thus, the insights from this study may facilitate the designing of pure antagonists or site II-mediated allosteric modulators to integrin αVβ3 to treat various diseases.
Collapse
Affiliation(s)
- Charles Mariasoosai
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Santanu Bose
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Senthil Natesan
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
13
|
Rodrigues Toledo C, Tantawy AA, Lima Fuscaldi L, Malavolta L, de Aguiar Ferreira C. EGFR- and Integrin α Vβ 3-Targeting Peptides as Potential Radiometal-Labeled Radiopharmaceuticals for Cancer Theranostics. Int J Mol Sci 2024; 25:8553. [PMID: 39126121 PMCID: PMC11313252 DOI: 10.3390/ijms25158553] [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: 06/26/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
The burgeoning field of cancer theranostics has witnessed advancements through the development of targeted molecular agents, particularly peptides. These agents exploit the overexpression or mutations of specific receptors, such as the Epidermal Growth Factor receptor (EGFR) and αVβ3 integrin, which are pivotal in tumor growth, angiogenesis, and metastasis. Despite the extensive research into and promising outcomes associated with antibody-based therapies, peptides offer a compelling alternative due to their smaller size, ease of modification, and rapid bioavailability, factors which potentially enhance tumor penetration and reduce systemic toxicity. However, the application of peptides in clinical settings has challenges. Their lower binding affinity and rapid clearance from the bloodstream compared to antibodies often limit their therapeutic efficacy and diagnostic accuracy. This overview sets the stage for a comprehensive review of the current research landscape as it relates to EGFR- and integrin αVβ3-targeting peptides. We aim to delve into their synthesis, radiolabeling techniques, and preclinical and clinical evaluations, highlighting their potential and limitations in cancer theranostics. This review not only synthesizes the extant literature to outline the advancements in peptide-based agents targeting EGFR and integrin αVβ3 but also identifies critical gaps that could inform future research directions. By addressing these gaps, we contribute to the broader discourse on enhancing the diagnostic precision and therapeutic outcomes of cancer treatments.
Collapse
Affiliation(s)
- Cibele Rodrigues Toledo
- The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA; (C.R.T.); (A.A.T.)
| | - Ahmed A. Tantawy
- The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA; (C.R.T.); (A.A.T.)
- Comparative Medicine and Integrative Biology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Leonardo Lima Fuscaldi
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo 01221-020, Brazil; (L.L.F.); (L.M.)
| | - Luciana Malavolta
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo 01221-020, Brazil; (L.L.F.); (L.M.)
| | - Carolina de Aguiar Ferreira
- The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA; (C.R.T.); (A.A.T.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
14
|
Zhao X, Jakobsson V, Tao Y, Zhao T, Wang J, Khong PL, Chen X, Zhang J. Targeted Radionuclide Therapy in Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39042829 DOI: 10.1021/acsami.4c07850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Despite the development of various novel therapies, glioblastoma (GBM) remains a devastating disease, with a median survival of less than 15 months. Recently, targeted radionuclide therapy has shown significant progress in treating solid tumors, with the approval of Lutathera for neuroendocrine tumors and Pluvicto for prostate cancer by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA). This achievement has shed light on the potential of targeted radionuclide therapy for other solid tumors, including GBM. This review presents the current status of targeted radionuclide therapy in GBM, highlighting the commonly used therapeutic radionuclides emitting alpha, beta particles, and Auger electrons that could induce potent molecular and cellular damage to treat GBM. We then explore a range of targeting vectors, including small molecules, peptides, and antibodies, which selectively target antigen-expressing tumor cells with minimal or no binding to healthy tissues. Considering that radiopharmaceuticals for GBM are often administered locoregionally to bypass the blood-brain barrier (BBB), we review prominent delivery methods such as convection-enhanced delivery, local implantation, and stereotactic injections. Finally, we address the challenges of this therapeutic approach for GBM and propose potential solutions.
Collapse
Affiliation(s)
- Xiaobin Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Vivianne Jakobsson
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yucen Tao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Tianzhi Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jingyan Wang
- Xiamen University, School of Public Health, Xiang'an South Road, Xiamen 361102, China
| | - Pek-Lan Khong
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Departments of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
15
|
Branco F, Cunha J, Mendes M, Vitorino C, Sousa JJ. Peptide-Hitchhiking for the Development of Nanosystems in Glioblastoma. ACS NANO 2024; 18:16359-16394. [PMID: 38861272 PMCID: PMC11223498 DOI: 10.1021/acsnano.4c01790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Glioblastoma (GBM) remains the epitome of aggressiveness and lethality in the spectrum of brain tumors, primarily due to the blood-brain barrier (BBB) that hinders effective treatment delivery, tumor heterogeneity, and the presence of treatment-resistant stem cells that contribute to tumor recurrence. Nanoparticles (NPs) have been used to overcome these obstacles by attaching targeting ligands to enhance therapeutic efficacy. Among these ligands, peptides stand out due to their ease of synthesis and high selectivity. This article aims to review single and multiligand strategies critically. In addition, it highlights other strategies that integrate the effects of external stimuli, biomimetic approaches, and chemical approaches as nanocatalytic medicine, revealing their significant potential in treating GBM with peptide-functionalized NPs. Alternative routes of parenteral administration, specifically nose-to-brain delivery and local treatment within the resected tumor cavity, are also discussed. Finally, an overview of the significant obstacles and potential strategies to overcome them are discussed to provide a perspective on this promising field of GBM therapy.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Joana Cunha
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria Mendes
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - João J. Sousa
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
16
|
Hovis G, Chandra N, Kejriwal N, Hsieh KJY, Chu A, Yang I, Wadehra M. Understanding the Role of Endothelial Cells in Glioblastoma: Mechanisms and Novel Treatments. Int J Mol Sci 2024; 25:6118. [PMID: 38892305 PMCID: PMC11173095 DOI: 10.3390/ijms25116118] [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: 05/09/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma is a highly aggressive neoplasm and the most common primary malignant brain tumor. Endothelial tissue plays a critical role in glioblastoma growth and progression, facilitating angiogenesis, cellular communication, and tumorigenesis. In this review, we present an up-to-date and comprehensive summary of the role of endothelial cells in glioblastomas, along with an overview of recent developments in glioblastoma therapies and tumor endothelial marker identification.
Collapse
Affiliation(s)
- Gabrielle Hovis
- Department of Neurosurgery, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Neha Chandra
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA (K.J.-Y.H.)
| | - Nidhi Kejriwal
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA (K.J.-Y.H.)
| | - Kaleb Jia-Yi Hsieh
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA (K.J.-Y.H.)
| | - Alison Chu
- Division of Neonatology and Developmental Biology, Department of Pediatrics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Isaac Yang
- Department of Neurosurgery, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Department of Radiation Oncology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Lundquist Institute, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Madhuri Wadehra
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA (K.J.-Y.H.)
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
17
|
Pan Y, Dang H, Zhou H, Fu H, Wu S, Liu H, Zhang J, Wang R, Tian Y, Xu B. A comparison study of dynamic [ 18F]Alfatide II imaging and [ 11C]MET in orthotopic rat models of glioblastoma. J Cancer Res Clin Oncol 2024; 150:208. [PMID: 38647690 PMCID: PMC11035414 DOI: 10.1007/s00432-024-05688-4] [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: 01/10/2024] [Accepted: 03/05/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE To investigate and compare the dynamic positron emission tomography (PET) imaging with [18F]Alfatide II Imaging and [11C]Methionine ([11C]MET) in orthotopic rat models of glioblastoma multiforme (GBM), and to assess the utility of [18F]Alfatide II in detecting and evaluating neoangiogenesis in GBM. METHODS [18F]Alfatide II and [11C]MET were injected into the orthotopic GBM rat models (n = 20, C6 glioma cells), followed by dynamic PET/MR scans 21 days after surgery of tumor implantation. On the PET image with both radiotracers, the MRI-based volume-of-interest (VOI) was manually delineated encompassing glioblastoma. Time-activity curves were expressed as tumor-to-normal brain ratio (TNR) parameters and PET pharmacokinetic modeling (PKM) performed using 2-tissue-compartment models (2TCM). Immunofluorescent staining (IFS), western blotting and blocking experiment of tumor tissue were performed for the validation. RESULTS Compared to 11C-MET, [18F]Alfatide II presented a persistent accumulation in the tumor, albeit with a slightly lower SUVmean of 0.79 ± 0.25, and a reduced uptake in the contralateral normal brain tissue, respectively. This resulted in a markedly higher tumor-to-normal brain ratio (TNR) of 18.22 ± 1.91. The time-activity curve (TACs) showed a significant increase in radioactive uptake in tumor tissue, followed by a plateau phase up to 60 min for [18F]Alfatide II (time to peak:255 s) and 40 min for [11C]MET (time to peak:135 s) post injection. PKM confirmed significantly higher K1 (0.23/0.07) and K3 (0.26/0.09) in the tumor region compared to the normal brain with [18F]Alfatide II. Compared to [11C]MET imaging, PKM confirmed both significantly higher K1/K2 (1.24 ± 0.79/1.05 ± 0.39) and K3/K4 (11.93 ± 4.28/3.89 ± 1.29) in the tumor region with [18F]Alfatide II. IFS confirmed significant expression of integrin and tumor vascularization in tumor region. CONCLUSION [18F]Alfatide II demonstrates potential in imaging tumor-associated neovascularization in the context of glioblastoma multiforme (GBM), suggesting its utility as a tool for further exploration in neovascular characterization.
Collapse
Affiliation(s)
- Yue Pan
- Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Haodan Dang
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Haoxi Zhou
- Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Huaping Fu
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Shina Wu
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Huanhuan Liu
- Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Jinming Zhang
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Ruimin Wang
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China
| | - Yuan Tian
- Department of Radiology, The 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Baixuan Xu
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, 100853, China.
| |
Collapse
|
18
|
Echavidre W, Durivault J, Gotorbe C, Blanchard T, Pagnuzzi M, Vial V, Raes F, Broisat A, Villeneuve R, Amblard R, Garnier N, Ortholan C, Faraggi M, Serrano B, Picco V, Montemagno C. Integrin-αvβ3 is a Therapeutically Targetable Fundamental Factor in Medulloblastoma Tumorigenicity and Radioresistance. CANCER RESEARCH COMMUNICATIONS 2023; 3:2483-2496. [PMID: 38009896 PMCID: PMC10702273 DOI: 10.1158/2767-9764.crc-23-0298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/05/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
Medulloblastoma is one of the most prevalent solid tumors found in children, occurring in the brain's posterior fossa. The standard treatment protocol involves maximal resection surgery followed by craniospinal irradiation and chemotherapy. Despite a long-term survival rate of 70%, wide disparities among patients have been observed. The identification of pertinent targets for both initial and recurrent medulloblastoma cases is imperative. Both primary and recurrent medulloblastoma are marked by their aggressive infiltration into surrounding brain tissue, robust angiogenesis, and resistance to radiotherapy. While the significant role of integrin-αvβ3 in driving these characteristics has been extensively documented in glioblastoma, its impact in the context of medulloblastoma remains largely unexplored. Integrin-αvβ3 was found to be expressed in a subset of patients with medulloblastoma. We investigated the role of integrin-αvβ3 using medulloblastoma-derived cell lines with β3-subunit depletion or overexpression both in vitro and in vivo settings. By generating radioresistant medulloblastoma cell lines, we uncovered an increased integrin-αvβ3 expression, which correlated with increased susceptibility to pharmacologic integrin-αvβ3 inhibition with cilengitide, a competitive ligand mimetic. Finally, we conducted single-photon emission computed tomography (SPECT)/MRI studies on orthotopic models using a radiolabeled integrin-αvβ3 ligand (99mTc-RAFT-RGD). This innovative approach presents the potential for a novel predictive imaging technique in the realm of medulloblastoma. Altogether, our findings lay the foundation for employing SPECT/MRI to identify a specific subset of patients with medulloblastoma eligible for integrin-αvβ3-directed therapies. This breakthrough offers a pathway toward more targeted and effective interventions in the treatment of medulloblastoma. SIGNIFICANCE This study demonstrates integrin-αvβ3's fundamental role in medulloblastoma tumorigenicity and radioresistance and the effect of its expression on cilengitide functional activity.
Collapse
Affiliation(s)
- William Echavidre
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco
| | - Jérôme Durivault
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco
| | - Célia Gotorbe
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco
| | - Thays Blanchard
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco
| | - Marina Pagnuzzi
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco
| | - Valérie Vial
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco
| | - Florian Raes
- Université de Grenoble Alpes, INSERM, LRB, Grenoble, France
| | - Alexis Broisat
- Université de Grenoble Alpes, INSERM, LRB, Grenoble, France
| | - Rémy Villeneuve
- Medical Physics Department, Centre Hospitalier Princesse Grace, Monaco, Monaco
| | - Régis Amblard
- Medical Physics Department, Centre Hospitalier Princesse Grace, Monaco, Monaco
| | - Nicolas Garnier
- Medical Physics Department, Centre Hospitalier Princesse Grace, Monaco, Monaco
| | - Cécile Ortholan
- Radiotherapy Department, Centre Hospitalier Princesse Grace, Monaco, Monaco
| | - Marc Faraggi
- Nuclear Medicine Department, Centre Hospitalier Princesse Grace, Monaco, Monaco
| | - Benjamin Serrano
- Medical Physics Department, Centre Hospitalier Princesse Grace, Monaco, Monaco
| | - Vincent Picco
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco
| | | |
Collapse
|
19
|
Viktorsson K, Rieckmann T, Fleischmann M, Diefenhardt M, Hehlgans S, Rödel F. Advances in molecular targeted therapies to increase efficacy of (chemo)radiation therapy. Strahlenther Onkol 2023; 199:1091-1109. [PMID: 37041372 PMCID: PMC10673805 DOI: 10.1007/s00066-023-02064-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/19/2023] [Indexed: 04/13/2023]
Abstract
Recent advances in understanding the tumor's biology in line with a constantly growing number of innovative technologies have prompted characterization of patients' individual malignancies and may display a prerequisite to treat cancer at its patient individual tumor vulnerability. In recent decades, radiation- induced signaling and tumor promoting local events for radiation sensitization were explored in detail, resulting the development of novel molecular targets. A multitude of pharmacological, genetic, and immunological principles, including small molecule- and antibody-based targeted strategies, have been developed that are suitable for combined concepts with radiation (RT) or chemoradiation therapy (CRT). Despite a plethora of promising experimental and preclinical findings, however, so far, only a very limited number of clinical trials have demonstrated a better outcome and/or patient benefit when RT or CRT are combined with targeted agents. The current review aims to summarize recent progress in molecular therapies targeting oncogenic drivers, DNA damage and cell cycle response, apoptosis signaling pathways, cell adhesion molecules, hypoxia, and the tumor microenvironment to impact therapy refractoriness and to boost radiation response. In addition, we will discuss recent advances in nanotechnology, e.g., RNA technologies and protein-degrading proteolysis-targeting chimeras (PROTACs) that may open new and innovative ways to benefit from molecular-targeted therapy approaches with improved efficacy.
Collapse
Affiliation(s)
- Kristina Viktorsson
- Department of Oncology/Pathology, Karolinska Institutet, Visionsgatan 4, 17164, Solna, Sweden
| | - Thorsten Rieckmann
- Department of Radiation Oncology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Department of Otolaryngology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Maximilian Fleischmann
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University of Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Markus Diefenhardt
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University of Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Stephanie Hehlgans
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
- Frankfurt Cancer Institute (FCI), University of Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
- German Cancer Consortium (DKTK) partner site: Frankfurt, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
20
|
Echavidre W, Fagret D, Faraggi M, Picco V, Montemagno C. Recent Pre-Clinical Advancements in Nuclear Medicine: Pioneering the Path to a Limitless Future. Cancers (Basel) 2023; 15:4839. [PMID: 37835533 PMCID: PMC10572076 DOI: 10.3390/cancers15194839] [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: 08/21/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
The theranostic approach in oncology holds significant importance in personalized medicine and stands as an exciting field of molecular medicine. Significant achievements have been made in this field in recent decades, particularly in treating neuroendocrine tumors using 177-Lu-radiolabeled somatostatin analogs and, more recently, in addressing prostate cancer through prostate-specific-membrane-antigen targeted radionuclide therapy. The promising clinical results obtained in these indications paved the way for the further development of this approach. With the continuous discovery of new molecular players in tumorigenesis, the development of novel radiopharmaceuticals, and the potential combination of theranostics agents with immunotherapy, nuclear medicine is poised for significant advancements. The strategy of theranostics in oncology can be categorized into (1) repurposing nuclear medicine agents for other indications, (2) improving existing radiopharmaceuticals, and (3) developing new theranostics agents for tumor-specific antigens. In this review, we provide an overview of theranostic development and shed light on its potential integration into combined treatment strategies.
Collapse
Affiliation(s)
- William Echavidre
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| | - Daniel Fagret
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, CHU Grenoble Alpes, Inserm, 38000 Grenoble, France;
| | - Marc Faraggi
- Nuclear Medicine Department, Centre Hospitalier Princesse Grace, 98000 Monaco, Monaco;
| | - Vincent Picco
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| | - Christopher Montemagno
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| |
Collapse
|
21
|
Martins C, Sarmento B. Multi-ligand functionalized blood-to-tumor sequential targeting strategies in the field of glioblastoma nanomedicine. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1893. [PMID: 37186374 DOI: 10.1002/wnan.1893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 05/17/2023]
Abstract
Glioblastoma (GBM) is an unmet clinical need characterized by a standard of care (SOC) 5-year survival rate of only 5%, and a treatment mostly palliative. Significant hurdles in GBM therapies include an effective penetration of therapeutics through the brain protective barrier, namely the blood-brain barrier (BBB), and a successful therapeutic delivery to brain-invading tumor cells post-BBB crossing. These hurdles, along with the poor prognosis and critical heterogeneity of the disease, have shifted attention to treatment modalities with capacity to precisely and sequentially target (i) BBB cells, inducing blood-to-brain transport, and (ii) GBM cells, leading to a higher therapeutic accumulation at the tumor site. This sequential targeting allows therapeutic molecules to reach the brain parenchyma and compromise molecular processes that support tumor cell invasion. Besides improving formulation and pharmacokinetics constraints of drugs, nanomedicines offer the possibility of being surface functionalized with multiple possibilities of targeting ligands, while delivering the desired therapeutic cargos to the biological sites of interest. Targeting ligands exploit the site-specific expression or overexpression of specific molecules on BBB and GBM cells, triggering brain plus tumor transport. Since the efficacy of single-ligand functionalized nanomedicines is limited due to the GBM anatomical site (brain) and disease complexity, this review presents an overview of multi-ligand functionalized, BBB and GBM sequentially- and dual-targeted nanomedicines reported in literature over the last 10 years. The role of the BBB in GBM progression, treatment options, and the multiple possibilities of currently available targeting ligands will be summarized. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Cláudia Martins
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- IUCS-CESPU, Gandra, Portugal
| |
Collapse
|
22
|
Yalamarty SSK, Filipczak N, Li X, Subhan MA, Parveen F, Ataide JA, Rajmalani BA, Torchilin VP. Mechanisms of Resistance and Current Treatment Options for Glioblastoma Multiforme (GBM). Cancers (Basel) 2023; 15:cancers15072116. [PMID: 37046777 PMCID: PMC10093719 DOI: 10.3390/cancers15072116] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive form of brain cancer that is difficult to treat due to its resistance to both radiation and chemotherapy. This resistance is largely due to the unique biology of GBM cells, which can evade the effects of conventional treatments through mechanisms such as increased resistance to cell death and rapid regeneration of cancerous cells. Additionally, the blood–brain barrier makes it difficult for chemotherapy drugs to reach GBM cells, leading to reduced effectiveness. Despite these challenges, there are several treatment options available for GBM. The standard of care for newly diagnosed GBM patients involves surgical resection followed by concurrent chemoradiotherapy and adjuvant chemotherapy. Emerging treatments include immunotherapy, such as checkpoint inhibitors, and targeted therapies, such as bevacizumab, that attempt to attack specific vulnerabilities in GBM cells. Another promising approach is the use of tumor-treating fields, a type of electric field therapy that has been shown to slow the growth of GBM cells. Clinical trials are ongoing to evaluate the safety and efficacy of these and other innovative treatments for GBM, intending to improve with outcomes for patients.
Collapse
Affiliation(s)
- Satya Siva Kishan Yalamarty
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Xiang Li
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Farzana Parveen
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- Department of Pharmacy Services, DHQ Hospital, Jhang 35200, Pakistan
| | - Janaína Artem Ataide
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas 13083-871, Brazil
| | - Bharat Ashok Rajmalani
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Vladimir P. Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
23
|
Soni N, Ora M, Jena A, Rana P, Mangla R, Ellika S, Almast J, Puri S, Meyers SP. Amino Acid Tracer PET MRI in Glioma Management: What a Neuroradiologist Needs to Know. AJNR Am J Neuroradiol 2023; 44:236-246. [PMID: 36657945 PMCID: PMC10187808 DOI: 10.3174/ajnr.a7762] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/21/2022] [Indexed: 01/21/2023]
Abstract
PET with amino acid tracers provides additional insight beyond MR imaging into the biology of gliomas that can be used for initial diagnosis, delineation of tumor margins, planning of surgical and radiation therapy, assessment of residual tumor, and evaluation of posttreatment response. Hybrid PET MR imaging allows the simultaneous acquisition of various PET and MR imaging parameters in a single investigation with reduced scanning time and improved anatomic localization. This review aimed to provide neuroradiologists with a concise overview of the various amino acid tracers and a practical understanding of the clinical applications of amino acid PET MR imaging in glioma management. Future perspectives in newer advances, novel radiotracers, radiomics, and cost-effectiveness are also outlined.
Collapse
Affiliation(s)
- N Soni
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - M Ora
- Sanjay Gandhi Postgraduate Institute of Medical Sciences (M.O.), Lucknow, Uttar Pradesh, India
| | - A Jena
- Indraprastha Apollo Hospital (A.J., P.R.), New Delhi, India
| | - P Rana
- Indraprastha Apollo Hospital (A.J., P.R.), New Delhi, India
| | - R Mangla
- Upstate University Hospital (R.M.), Syracuse, New York
| | - S Ellika
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - J Almast
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - S Puri
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - S P Meyers
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| |
Collapse
|
24
|
Dadgar H, Jokar N, Nemati R, Larvie M, Assadi M. PET tracers in glioblastoma: Toward neurotheranostics as an individualized medicine approach. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1103262. [PMID: 39355049 PMCID: PMC11440984 DOI: 10.3389/fnume.2023.1103262] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/23/2023] [Indexed: 10/03/2024]
Abstract
Over the past decade, theragnostic radiopharmaceuticals have been used in nuclear medicine for both diagnosis and treatment of various tumors. In this review, we carried out a literature search to investigate and explain the role of radiotracers in the theragnostic approach to glioblastoma multiform (GBM). We primarily focused on basic and rather common positron emotion tomography (PET) radiotracers in these tumors. Subsequently, we introduced and evaluated the preclinical and clinical results of theranostic-based biomarkers including integrin receptor family, prostate-specific membrane antigen (PSMA), fibroblast activated protein (FAP), somatostatin receptors (SRS), and chemokine receptor-4 (CXCR4) for patients with GBM to confer the benefit of personalized therapy. Moreover, promising research opportunities that could have a profound impact on the treatment of GBM over the next decade are also highlighted. Preliminary results showed the potential feasibility of the theragnostic approach using theses biomarkers in GBM patients.
Collapse
Affiliation(s)
- Habibullah Dadgar
- Cancer Research Center, RAZAVI Hospital, Imam Reza International University, Mashhad, Iran
| | - Narges Jokar
- The Persian Gulf Nuclear Medicine Research Center, Department of Molecular Imaging and Theranostics, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Reza Nemati
- Department of Neurology, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mykol Larvie
- Department of Radiology, Cleveland Clinic, Cleveland, Ohio
| | - Majid Assadi
- The Persian Gulf Nuclear Medicine Research Center, Department of Molecular Imaging and Theranostics, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
25
|
Xia D, Zhang Y, Zhang C, Yao X, Tang Y, Wang F, Han X, Yin H, Xu C, Gao X. Observation of the protein expression level via naked eye: Pt clusters catalyze non-color molecules into brown-colored molecules in cells. Front Chem 2023; 11:1145415. [PMID: 36860645 PMCID: PMC9969140 DOI: 10.3389/fchem.2023.1145415] [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: 01/16/2023] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
α v β 3 is overexpressed in various tumor cells and plays a key role in tumor genesis, invasion, and metastasis. Therefore, it is of great significance to precisely detect the α v β 3 level in cells via a simple method. For this purpose, we have constructed a peptide-coated platinum (Pt) cluster. Due to its bright fluorescence, well-defined Pt atom numbers, and peroxidase-like catalytic activity, this cluster can be used to evaluate α v β 3 levels in cells by fluorescence imaging, inductively coupled plasma mass spectrometry (ICP-MS), and catalytic amplification of visual dyes, respectively. In this report, the expression level of α v β 3 in living cells is well-detected by the naked eye under an ordinary light microscope when the Pt cluster binds to αvβ3 in cells and catalyzes non-color 3,3'-diaminobenzidine (DAB) into brown-colored molecules in situ. Moreover, SiHa, HeLa, and 16HBE cell lines with different α v β 3 expression levels can be visually distinguished by the peroxidase-like Pt clusters. This research will provide a reliable method for the simple detection of α v β 3 levels in cells.
Collapse
Affiliation(s)
- Dongfang Xia
- College of Chemistry and Material Science, Shandong Agricultural University, Taian, Shandong, China
| | - Yong Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Chunyu Zhang
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiuxiu Yao
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Yuhua Tang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Fuchao Wang
- College of Chemistry and Material Science, Shandong Agricultural University, Taian, Shandong, China
| | - Xu Han
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Beijing, China
| | - Hongzong Yin
- College of Chemistry and Material Science, Shandong Agricultural University, Taian, Shandong, China,*Correspondence: Hongzong Yin, ; Chao Xu, ; Xueyun Gao,
| | - Chao Xu
- College of Chemistry and Material Science, Shandong Agricultural University, Taian, Shandong, China,*Correspondence: Hongzong Yin, ; Chao Xu, ; Xueyun Gao,
| | - Xueyun Gao
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China,*Correspondence: Hongzong Yin, ; Chao Xu, ; Xueyun Gao,
| |
Collapse
|
26
|
Bian Y, Wang Y, Chen X, Zhang Y, Xiong S, Su D. Image‐guided diagnosis and treatment of glioblastoma. VIEW 2023. [DOI: 10.1002/viw.20220069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Yongning Bian
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| | - Yaling Wang
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| | - Xueqian Chen
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| | - Yong Zhang
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| | - Shaoqing Xiong
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| | - Dongdong Su
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| |
Collapse
|
27
|
Kostryukova LV, Tereshkina YA, Tikhonova EG, Sanzhakov MA, Bobrova DV, Khudoklinova YY. [Study of the efficiency of cellular accumulation of doxorubicin supplied with a targeted delivery system based on phospholipid nanoparticles with integrin-directed peptide]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:437-443. [PMID: 36573410 DOI: 10.18097/pbmc20226806437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Chemotherapeutic agents containing targeted systems are a promising pathway to increase the effectiveness of glioblastoma treatment. Specific proteins characterized by increased expression on the surface of tumor cells are considered as possible targets. Integrin αvβ3 is one of such proteins on the cell surface. It effectively binds the cyclic Arg-Gly-Asp (cRGD) peptide. In this study, the cRGD peptide-modified doxorubicin (Dox) phospholipid composition was investigated. The particle size of this composition was 43.76±2.09 nm, the ζ-potential was 4.33±0.54 mV. Dox was almost completely incorporated into the nanoparticles (99.7±0.58%). The drug release increased in an acidic medium (at pH 5.0 of about 35±3.2%). The total accumulation and internalization of Dox used the composition of phospholipid nanoparticles with the targeted vector was 1.4-fold higher as compared to the free form. In the HeLa cell line (not expressing αvβ3 integrin) this effect was not observed. These results suggest the prospects of using the cyclic RGD peptide in the delivery of Dox to glioblastoma cells and the feasibility of further investigation of the mechanism of action of the entire composition as a whole.
Collapse
Affiliation(s)
| | | | | | | | - D V Bobrova
- Institute of Biomedical Chemistry, Moscow, Russia
| | | |
Collapse
|
28
|
Internalisation of RGD-Engineered Extracellular Vesicles by Glioblastoma Cells. BIOLOGY 2022; 11:biology11101483. [PMID: 36290387 PMCID: PMC9598886 DOI: 10.3390/biology11101483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Simple Summary Glioblastoma multiforme (GBM) is the most aggressive and malignant type of central nervous system (CNS) tumour. Although several treatment options are available, patients generally succumb within 14 months after diagnosis. With the rapid progression of exosome bioengineering technologies, novel therapy opportunities are emerging for GBM treatment. The surface of GBM cells is characterised by the overexpression of transmembrane receptor integrins, which are essential for cell interactions with several proteins in the extracellular matrix. Therefore, integrin-binding drug delivery vehicles have been proposed as a potential strategy for glioblastoma therapy. Small extracellular vesicles possess several attractive characteristics for drug delivery: small size, biocompatibility, ability to cross the blood–brain barrier and capacity to be loaded with exogenous materials. Current bioengineering technologies further increase extracellular vesicle capabilities by loading them with anticancer drugs and incorporating targeting ligands. This study explored the capacity of Arginylglycylaspartic acid (RGD, or Arginine–Glycine–Aspartate)-polypeptide-engineered extracellular vesicles to internalise and deliver loaded cargo in GBM cells. The results demonstrate that introducing the RGD ligand to extracellular vesicles could significantly increase their internalisation by GBM cells and hence improve drug delivery efficacy. Abstract Glioblastoma multiforme (GBM) is the most aggressive CNS tumour with no efficient treatment, partly due to the retention of anticancer drugs by the blood–brain barrier (BBB) and their insufficient concentration in tumour cells. Extracellular vesicles (EVs) are attractive drug carriers because of their biocompatibility and ability to cross the BBB. Additional efficiency can be achieved by adding GBM-cell-specific ligands. GBM cells overexpress integrins; thus, one of the most straightforward targeting strategies is to modify EVs with integrin-recognising molecules. This study investigated the therapeutic potential of genetically engineered EVs with elevated membrane levels of the integrin-binding peptide RGD (RGD-EVs) against GBM cells in vitro. For RGD-EV production, stable RGD-HEK 293FT cells were generated by using a pcDNA4/TO-Lamp2b-iRGD-HA expression vector and performing antibiotic-based selection. RGD-EVs were isolated from RGD-HEK 293FT-cell-conditioned medium and characterised by size (Zetasizer), specific markers (ELISA) and RGD expression (Western Blot). Internalisation by human GBM cells HROG36 and U87 MG and BJ-5ta human fibroblasts was assessed by fluorescent EV RNA labelling. The effect of doxorubicin-loaded RGD-EVs on GBM cells was evaluated by the metabolic PrestoBlue viability assay; functional GAPDH gene knockdown by RGD-EV-encapsulated siRNA was determined by RT-qPCR. RGD-EVs had 40% higher accumulation in GBM cells (but not in fibroblasts) and induced significantly stronger toxicity by loaded doxorubicin and GAPDH silencing by loaded siRNA compared to unmodified EVs. Thus, RGD modification substantially increases the specific delivery capacity of HEK 293FT-derived EVs to GBM cells.
Collapse
|
29
|
Identification of Prognostic Genes in Gliomas Based on Increased Microenvironment Stiffness. Cancers (Basel) 2022; 14:cancers14153659. [PMID: 35954323 PMCID: PMC9367320 DOI: 10.3390/cancers14153659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
With a median survival time of 15 months, glioblastoma multiforme is one of the most aggressive primary brain cancers. The crucial roles played by the extracellular matrix (ECM) stiffness in glioma progression and treatment resistance have been reported in numerous studies. However, the association between ECM-stiffness-regulated genes and the prognosis of glioma patients remains to be explored. Thus, using bioinformatics analysis, we first identified 180 stiffness-dependent genes from an RNA-Seq dataset, and then evaluated their prognosis in The Cancer Genome Atlas (TCGA) glioma dataset. Our results showed that 11 stiffness-dependent genes common between low- and high-grade gliomas were prognostic. After validation using the Chinese Glioma Genome Atlas (CGGA) database, we further identified four stiffness-dependent prognostic genes: FN1, ITGA5, OSMR, and NGFR. In addition to high-grade glioma, overexpression of the four-gene signature also showed poor prognosis in low-grade glioma patients. Moreover, our analysis confirmed that the expression levels of stiffness-dependent prognostic genes in high-grade glioma were significantly higher than in low-grade glioma, suggesting that these genes were associated with glioma progression. Based on a pathophysiology-inspired approach, our findings illuminate the link between ECM stiffness and the prognosis of glioma patients and suggest a signature of four stiffness-dependent genes as potential therapeutic targets.
Collapse
|