1
|
Jiménez R, Constantinescu A, Yazir M, Alfonso-Triguero P, Pequerul R, Parés X, Pérez-Alea M, Candiota AP, Farrés J, Lorenzo J. Targeting Retinaldehyde Dehydrogenases to Enhance Temozolomide Therapy in Glioblastoma. Int J Mol Sci 2024; 25:11512. [PMID: 39519068 PMCID: PMC11546810 DOI: 10.3390/ijms252111512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma (GB) is an aggressive malignant central nervous system tumor that is currently incurable. One of the main pitfalls of GB treatment is resistance to the chemotherapeutic standard of care, temozolomide (TMZ). The role of aldehyde dehydrogenases (ALDHs) in the glioma stem cell (GSC) subpopulation has been related to chemoresistance. ALDHs take part in processes such as cell proliferation, differentiation, invasiveness or metastasis and have been studied as pharmacological targets in cancer treatment. In the present work, three novel α,β-acetylenic amino thiolester compounds, with demonstrated efficacy as ALDH inhibitors, were tested in vitro on a panel of six human GB cell lines and one murine GB cell line. Firstly, the expression of the ALDH1A isoforms was assessed, and then inhibitors were tested for their cytotoxicity and their ability to inhibit cellular ALDH activity. Drug combination assays with TMZ were performed, as well as an assessment of the cell death mechanism and generation of ROS. A knockout of several ALDH genes was carried out in one of the human GB cell lines, allowing us to discuss their role in cell proliferation, migration capacity and resistance to treatment. Our results strongly suggest that ALDH inhibitors could be an interesting approach in the treatment of GB, with EC50 values in the order of micromolar, decreasing ALDH activity in GB cell lines to 40-50%.
Collapse
Affiliation(s)
- Rafael Jiménez
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain; (R.J.); (P.A.-T.); (R.P.); (X.P.); (A.P.C.)
- Institute for Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
| | - Andrada Constantinescu
- Unit of Research in Cellular and Molecular Biology, Advanced BioDesign, Saint-Priest, 69800 Lyon, France; (A.C.); (M.Y.); (M.P.-A.)
| | - Muhube Yazir
- Unit of Research in Cellular and Molecular Biology, Advanced BioDesign, Saint-Priest, 69800 Lyon, France; (A.C.); (M.Y.); (M.P.-A.)
| | - Paula Alfonso-Triguero
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain; (R.J.); (P.A.-T.); (R.P.); (X.P.); (A.P.C.)
- Institute for Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, E-08193 Bellaterra, Spain
| | - Raquel Pequerul
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain; (R.J.); (P.A.-T.); (R.P.); (X.P.); (A.P.C.)
- Unit of Research in Cellular and Molecular Biology, Advanced BioDesign, Saint-Priest, 69800 Lyon, France; (A.C.); (M.Y.); (M.P.-A.)
| | - Xavier Parés
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain; (R.J.); (P.A.-T.); (R.P.); (X.P.); (A.P.C.)
| | - Mileidys Pérez-Alea
- Unit of Research in Cellular and Molecular Biology, Advanced BioDesign, Saint-Priest, 69800 Lyon, France; (A.C.); (M.Y.); (M.P.-A.)
| | - Ana Paula Candiota
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain; (R.J.); (P.A.-T.); (R.P.); (X.P.); (A.P.C.)
- Institute for Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, E-08913 Bellaterra, Spain
| | - Jaume Farrés
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain; (R.J.); (P.A.-T.); (R.P.); (X.P.); (A.P.C.)
| | - Julia Lorenzo
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain; (R.J.); (P.A.-T.); (R.P.); (X.P.); (A.P.C.)
- Institute for Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, E-08913 Bellaterra, Spain
| |
Collapse
|
2
|
Duenas-Gonzalez A, Gonzalez-Fierro A, Bornstein-Quevedo L, Gutierrez-Delgado F, Kast RE, Chavez-Blanco A, Dominguez-Gomez G, Candelaria M, Romo-Pérez A, Correa-Basurto J, Lizano M, Perez-de la Cruz V, Robles-Bañuelos B, Nuñez-Corona D, Martinez-Perez E, Verastegui E. Multitargeted polypharmacotherapy for cancer treatment. theoretical concepts and proposals. Expert Rev Anticancer Ther 2024; 24:665-677. [PMID: 38913911 DOI: 10.1080/14737140.2024.2372336] [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: 01/26/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION The pharmacological treatment of cancer has evolved from cytotoxic to molecular targeted therapy. The median survival gains of 124 drugs approved by the FDA from 2003 to 2021 is 2.8 months. Targeted therapy is based on the somatic mutation theory, which has some paradoxes and limitations. While efforts of targeted therapy must continue, we must study newer approaches that could advance therapy and affordability for patients. AREAS COVERED This work briefly overviews how cancer therapy has evolved from cytotoxic chemotherapy to current molecular-targeted therapy. The limitations of the one-target, one-drug approach considering cancer as a robust system and the basis for multitargeting approach with polypharmacotherapy using repurposing drugs. EXPERT OPINION Multitargeted polypharmacotherapy for cancer with repurposed drugs should be systematically investigated in preclinical and clinical studies. Remarkably, most of these proposed drugs already have a long history in the clinical setting, and their safety is known. In principle, the risk of their simultaneous administration should not be greater than that of a first-in-human phase I study as long as the protocol is developed with strict vigilance to detect early possible side effects from their potential interactions. Research on cancer therapy should go beyond the prevailing paradigm targeted therapy.
Collapse
Affiliation(s)
- Alfonso Duenas-Gonzalez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas UNAM, Mexico City, Mexico
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Aurora Gonzalez-Fierro
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | - Francisco Gutierrez-Delgado
- Centro de Estudios y Prevención del Cancer Tuxtla Gutiérrez, Chiapas, México; Latin American School of Oncology (ELO), México City, Mexico
| | - Richard E Kast
- Head of Faculty, Brain Study, IIAIG Study Center, Burlington, VT, USA
| | - Alma Chavez-Blanco
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | - Myrna Candelaria
- Departamento de Hematología, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Adriana Romo-Pérez
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jose Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, SEPI-ESM, Instituto Politécnico Nacional, México, Mexico City, Mexico
| | - Marcela Lizano
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas UNAM, Mexico City, Mexico
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Veronica Perez-de la Cruz
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City, Mexico
| | | | - David Nuñez-Corona
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Erandi Martinez-Perez
- Subdireccion de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Emma Verastegui
- Departamento de Cuidados Paliativos, Division de Cirugia, Instituto Nacional de Cancerologia, Mexico City, Mexico
| |
Collapse
|
3
|
Chantzi E, Hammerling U, Gustafsson MG. Exhaustive in vitro evaluation of the 9-drug cocktail CUSP9 for treatment of glioblastoma. Comput Biol Med 2024; 178:108748. [PMID: 38925084 DOI: 10.1016/j.compbiomed.2024.108748] [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: 11/28/2023] [Revised: 05/23/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024]
Abstract
The CUSP9 protocol is a polypharmaceutical strategy aiming at addressing the complexity of glioblastoma by targeting multiple pathways. Although the rationale for this 9-drug cocktail is well-supported by theoretical and in vitro data, its effectiveness compared to its 511 possible subsets has not been comprehensively evaluated. Such an analysis could reveal if fewer drugs could achieve similar or better outcomes. We conducted an exhaustive in vitro evaluation of the CUSP9 protocol using COMBImageDL, our specialized framework for testing higher-order drug combinations. This study assessed all 511 subsets of the CUSP9v3 protocol, in combination with temozolomide, on two clonal cultures of glioma-initiating cells derived from patient samples. The drugs were used at fixed, clinically relevant concentrations, and the experiment was performed in quadruplicate with endpoint cell viability and live-cell imaging readouts. Our results showed that several lower-order drug combinations produced effects equivalent to the full CUSP9 cocktail, indicating potential for simplified regimens in personalized therapy. Further validation through in vivo and precision medicine testing is required. Notably, a subset of four drugs (auranofin, disulfiram, itraconazole, sertraline) was particularly effective, reducing cell growth, altering cell morphology, increasing apoptotic-like cells within 4-28 h, and significantly decreasing cell viability after 68 h compared to untreated cells. This study underscores the importance and feasibility of comprehensive in vitro evaluations of complex drug combinations on patient-derived tumor cells, serving as a critical step toward (pre-)clinical development.
Collapse
Affiliation(s)
- Efthymia Chantzi
- Department of Medical Sciences, Cancer Pharmacology and Computational Medicine, Uppsala University, Sweden.
| | - Ulf Hammerling
- Department of Civil & Industrial Engineering, Industrial Analytics, Uppsala University, Sweden
| | - Mats G Gustafsson
- Department of Civil & Industrial Engineering, Industrial Analytics, Uppsala University, Sweden.
| |
Collapse
|
4
|
Kast RE. IC Regimen: Delaying Resistance to Lorlatinib in ALK Driven Cancers by Adding Repurposed Itraconazole and Cilostazol. Cells 2024; 13:1175. [PMID: 39056757 PMCID: PMC11274432 DOI: 10.3390/cells13141175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Lorlatinib is a pharmaceutical ALK kinase inhibitor used to treat ALK driven non-small cell lung cancers. This paper analyses the intersection of past published data on the physiological consequences of two unrelated drugs from general medical practice-itraconazole and cilostazol-with the pathophysiology of ALK positive non-small cell lung cancer. A conclusion from that data analysis is that adding itraconazole and cilostazol may make lorlatinib more effective. Itraconazole, although marketed worldwide as a generic antifungal drug, also inhibits Hedgehog signaling, Wnt signaling, hepatic CYP3A4, and the p-gp efflux pump. Cilostazol, marketed worldwide as a generic thrombosis preventative drug, acts by inhibiting phosphodiesterase 3, and, by so doing, lowers platelets' adhesion, thereby partially depriving malignant cells of the many tumor trophic growth factors supplied by platelets. Itraconazole may enhance lorlatinib effectiveness by (i) reducing or stopping a Hedgehog-ALK amplifying feedback loop, by (ii) increasing lorlatinib's brain levels by p-gp inhibition, and by (iii) inhibiting growth drive from Wnt signaling. Cilostazol, surprisingly, carries minimal bleeding risk, lower than that of aspirin. Risk/benefit assessment of the combination of metastatic ALK positive lung cancer being a low-survival disease with the predicted safety of itraconazole-cilostazol augmentation of lorlatinib favors a trial of this drug trio in ALK positive lung cancer.
Collapse
|
5
|
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
|
6
|
Cao Q, Hajosch A, Kast RE, Loehmann C, Hlavac M, Fischer-Posovszky P, Strobel H, Westhoff MA, Siegelin MD, Wirtz CR, Halatsch ME, Karpel-Massler G. Tumor Treating Fields (TTFields) combined with the drug repurposing approach CUSP9v3 induce metabolic reprogramming and synergistic anti-glioblastoma activity in vitro. Br J Cancer 2024; 130:1365-1376. [PMID: 38396172 PMCID: PMC11015043 DOI: 10.1038/s41416-024-02608-8] [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: 06/18/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Glioblastoma represents a brain tumor with a notoriously poor prognosis. First-line therapy may include adjunctive Tumor Treating Fields (TTFields) which are electric fields that are continuously delivered to the brain through non-invasive arrays. On a different note, CUSP9v3 represents a drug repurposing strategy that includes 9 repurposed drugs plus metronomic temozolomide. Here, we examined whether TTFields enhance the antineoplastic activity of CUSP9v3 against this disease. METHODS We performed preclinical testing of a multimodal approach of TTFields and CUSP9v3 in different glioblastoma models. RESULTS TTFields had predominantly synergistic inhibitory effects on the cell viability of glioblastoma cells and non-directed movement was significantly impaired when combined with CUSP9v3. TTFields plus CUSP9v3 significantly enhanced apoptosis, which was associated with a decreased mitochondrial outer membrane potential (MOMP), enhanced cleavage of effector caspase 3 and reduced expression of Bcl-2 and Mcl-1. Moreover, oxidative phosphorylation and expression of respiratory chain complexes I, III and IV was markedly reduced. CONCLUSION TTFields strongly enhance the CUSP9v3-mediated anti-glioblastoma activity. TTFields are currently widely used for the treatment of glioblastoma patients and CUSP9v3 was shown to have a favorable safety profile in a phase Ib/IIa trial (NCT02770378) which facilitates transition of this multimodal approach to the clinical setting.
Collapse
Affiliation(s)
- Qiyu Cao
- Department of Neurosurgery, Ulm University Medical Center, Ulm, Germany
| | - Annika Hajosch
- Department of Neurosurgery, Ulm University Medical Center, Ulm, Germany
| | | | | | - Michal Hlavac
- Department of Neurosurgery, Ulm University Medical Center, Ulm, Germany
| | | | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Markus D Siegelin
- Department of Pathology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Marc-Eric Halatsch
- Department of Neurosurgery, Cantonal Hospital of Winterthur, Winterthur, Switzerland
| | | |
Collapse
|
7
|
Halatsch ME. Special Issue: Principal Challenges in the Adjuvant Treatment of Glioblastoma. Biomedicines 2023; 11:1881. [PMID: 37509520 PMCID: PMC10377107 DOI: 10.3390/biomedicines11071881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 06/13/2023] [Indexed: 07/30/2023] Open
Abstract
Despite advances in local treatments, such as supramaximal resection (even in eloquent locations [...].
Collapse
Affiliation(s)
- Marc-Eric Halatsch
- Department of Neurosurgery, Cantonal Hospital of Winterthur, CH-8400 Winterthur, Switzerland
| |
Collapse
|
8
|
Boylan J, Byers E, Kelly DF. The Glioblastoma Landscape: Hallmarks of Disease, Therapeutic Resistance, and Treatment Opportunities. MEDICAL RESEARCH ARCHIVES 2023; 11:10.18103/mra.v11i6.3994. [PMID: 38107346 PMCID: PMC10723753 DOI: 10.18103/mra.v11i6.3994] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Malignant brain tumors are aggressive and difficult to treat. Glioblastoma is the most common and lethal form of primary brain tumor, often found in patients with no genetic predisposition. The median life expectancy for individuals diagnosed with this condition is 6 months to 2 years and there is no known cure. New paradigms in cancer biology implicate a small subset of tumor cells in initiating and sustaining these incurable brain tumors. Here, we discuss the heterogenous nature of glioblastoma and theories behind its capacity for therapy resistance and recurrence. Within the cancer landscape, cancer stem cells are thought to be both tumor initiators and major contributors to tumor heterogeneity and therapy evasion and such cells have been identified in glioblastoma. At the cellular level, disruptions in the delicate balance between differentiation and self-renewal spur transformation and support tumor growth. While rapidly dividing cells are more sensitive to elimination by traditional treatments, glioblastoma stem cells evade these measures through slow division and reversible exit from the cell cycle. At the molecular level, glioblastoma tumor cells exploit several signaling pathways to evade conventional therapies through improved DNA repair mechanisms and a flexible state of senescence. We examine these common evasion techniques while discussing potential molecular approaches to better target these deadly tumors. Equally important, the presented information encourages the idea of augmenting conventional treatments with novel glioblastoma stem cell-directed therapies, as eliminating these harmful progenitors holds great potential to modulate tumor recurrence.
Collapse
Affiliation(s)
- Jack Boylan
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
- Center for Structural Oncology, Pennsylvania State University, University Park, PA 16802, USA
- Molecular, Cellular, and Integrative Biosciences Graduate Program, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Elizabeth Byers
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
- Molecular, Cellular, and Integrative Biosciences Graduate Program, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Deborah F. Kelly
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
- Center for Structural Oncology, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
9
|
Manfreda L, Rampazzo E, Persano L. Wnt Signaling in Brain Tumors: A Challenging Therapeutic Target. BIOLOGY 2023; 12:biology12050729. [PMID: 37237541 DOI: 10.3390/biology12050729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023]
Abstract
The involvement of Wnt signaling in normal tissue homeostasis and disease has been widely demonstrated over the last 20 years. In particular, dysregulation of Wnt pathway components has been suggested as a relevant hallmark of several neoplastic malignancies, playing a role in cancer onset, progression, and response to treatments. In this review, we summarize the current knowledge on the instructions provided by Wnt signaling during organogenesis and, particularly, brain development. Moreover, we recapitulate the most relevant mechanisms through which aberrant Wnt pathway activation may impact on brain tumorigenesis and brain tumor aggressiveness, with a particular focus on the mutual interdependency existing between Wnt signaling components and the brain tumor microenvironment. Finally, the latest anti-cancer therapeutic approaches employing the specific targeting of Wnt signaling are extensively reviewed and discussed. In conclusion, here we provide evidence that Wnt signaling, due to its pleiotropic involvement in several brain tumor features, may represent a relevant target in this context, although additional efforts will be needed to: (i) demonstrate the real clinical impact of Wnt inhibition in these tumors; (ii) overcome some still unsolved concerns about the potential systemic effects of such approaches; (iii) achieve efficient brain penetration.
Collapse
Affiliation(s)
- Lorenzo Manfreda
- Department of Women and Children's Health, University of Padova, Via Giustininani, 3, 35128 Padova, Italy
- Pediatric Research Institute, Corso Stati Uniti, 4, 35127 Padova, Italy
| | - Elena Rampazzo
- Department of Women and Children's Health, University of Padova, Via Giustininani, 3, 35128 Padova, Italy
- Pediatric Research Institute, Corso Stati Uniti, 4, 35127 Padova, Italy
| | - Luca Persano
- Department of Women and Children's Health, University of Padova, Via Giustininani, 3, 35128 Padova, Italy
- Pediatric Research Institute, Corso Stati Uniti, 4, 35127 Padova, Italy
| |
Collapse
|
10
|
Benkő BM, Lamprou DA, Sebestyén A, Zelkó R, Sebe I. Clinical, pharmacological, and formulation evaluation of disulfiram in the treatment of glioblastoma - a systematic literature review. Expert Opin Drug Deliv 2023; 20:541-557. [PMID: 36922013 DOI: 10.1080/17425247.2023.2190581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
INTRODUCTION Glioblastoma (GB) is one of the most challenging central nervous system (CNS) tumors in treatment options and response, urging the development of novel management strategies. The anti-alcoholism drug, disulfiram (DS), has a potential anticancer activity, and its complex mechanism of action is assumed to be well exploited against the heterogeneous GB. AREA COVERED Through a systematic literature review about repositioning DS to GB treatment, an evaluation of the clinical, pharmacological, and formulation strategies is provided to specify the challenges of drug delivery and thus to advance its clinical translation. From six databases, 35 articles were selected, including case report (1); clinical trials (3); original articles mainly representing in vitro and preclinical pharmacological data, and 10 dealing with technological approaches. EXPERT OPINION The repositioning of DS in GB treatment is facing drug and tumor-associated limitations due to the oral drug's low bioavailability, unwanted metabolism, and inefficient delivery to brain-tumor tissue. Development strategies using molecular encapsulation of DS and the parenteral dosage forms improve the anticancer pharmacology of the drug. The development of optimized drug delivery systems (DDS) shows promise for the clinical translation of DS into GB adjuvant therapy.
Collapse
Affiliation(s)
- Beáta-Mária Benkő
- University Pharmacy Department of Pharmacy Administration, Semmelweis University, Budapest, Hungary
| | | | - Anna Sebestyén
- Tumour Biology, Cell and Tissue Culture Laboratory, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Romána Zelkó
- University Pharmacy Department of Pharmacy Administration, Semmelweis University, Budapest, Hungary
| | - István Sebe
- University Pharmacy Department of Pharmacy Administration, Semmelweis University, Budapest, Hungary
| |
Collapse
|
11
|
El Atat O, Naser R, Abdelkhalek M, Habib RA, El Sibai M. Molecular targeted therapy: A new avenue in glioblastoma treatment. Oncol Lett 2022; 25:46. [PMID: 36644133 PMCID: PMC9811647 DOI: 10.3892/ol.2022.13632] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/21/2022] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma, also referred to as glioblastoma multiforme (GBM), is grade IV astrocytoma characterized by being fast-growing and the most aggressive brain tumor. In adults, it is the most prevalent type of malignant brain tumor. Despite the advancements in both diagnosis tools and therapeutic treatments, GBM is still associated with poor survival rate without any statistically significant improvement in the past three decades. Patient's genome signature is one of the key factors causing the development of this tumor, in addition to previous radiation exposure and other environmental factors. Researchers have identified genomic and subsequent molecular alterations affecting core pathways that trigger the malignant phenotype of this tumor. Targeting intrinsically altered molecules and pathways is seen as a novel avenue in GBM treatment. The present review shed light on signaling pathways and intrinsically altered molecules implicated in GBM development. It discussed the main challenges impeding successful GBM treatment, such as the blood brain barrier and tumor microenvironment (TME), the plasticity and heterogeneity of both GBM and TME and the glioblastoma stem cells. The present review also presented current advancements in GBM molecular targeted therapy in clinical trials. Profound and comprehensive understanding of molecular participants opens doors for innovative, more targeted and personalized GBM therapeutic modalities.
Collapse
Affiliation(s)
- Oula El Atat
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Rayan Naser
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Maya Abdelkhalek
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Ralph Abi Habib
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Mirvat El Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon,Correspondence to: Professor Mirvat El Sibai, Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Koraytem Street, Beirut 1102 2801, Lebanon, E-mail:
| |
Collapse
|
12
|
Konig S, Strobel H, Grunert M, Lyszkiewicz M, Brühl O, Karpel-Massler G, Ziętara N, La Ferla-Brühl K, Siegelin MD, Debatin KM, Westhoff MA. Unblinding the watchmaker: cancer treatment and drug design in the face of evolutionary pressure. Expert Opin Drug Discov 2022; 17:1081-1094. [PMID: 35997138 DOI: 10.1080/17460441.2022.2114454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Death due to cancer is mostly associated with therapy ineffectiveness, i.e. tumor cells no longer responding to treatment. The underlying dynamics that facilitate this mutational escape from selective pressure are well studied in several other fields and several interesting approaches exist to combat this phenomenon, for example in the context of antibiotic-resistance in bacteria. AREAS COVERED Ninety percent of all cancer-related deaths are associated with treatment failure. Here, we discuss the common treatment modalities and prior attempts to overcome acquired resistance to therapy. The underlying molecular mechanisms are discussed and the implications of emerging resistance in other systems, such as bacteria, are discussed in the context of cancer. EXPERT OPINION Reevaluating emerging therapy resistance in tumors as an evolutionary mechanism to survive in a rapidly and drastically altering fitness landscape leads to novel treatment strategies and distinct requirements for new drugs. Here, we propose a scheme of considerations that need to be applied prior to the discovery of novel therapeutic drugs.
Collapse
Affiliation(s)
- Sophia Konig
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| | - Michael Grunert
- Department of Nuclear Medicine, German Armed Forces Hospital of Ulm, Ulm, Germany
| | - Marcin Lyszkiewicz
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| | - Oliver Brühl
- Laboratorio Analisi Sicilia, Catania, Lentini, Italy
| | | | - Natalia Ziętara
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Germany
| | | | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, Albany, NY, USA
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
13
|
Ntafoulis I, Koolen SLW, Leenstra S, Lamfers MLM. Drug Repurposing, a Fast-Track Approach to Develop Effective Treatments for Glioblastoma. Cancers (Basel) 2022; 14:3705. [PMID: 35954371 PMCID: PMC9367381 DOI: 10.3390/cancers14153705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/10/2022] Open
Abstract
Glioblastoma (GBM) remains one of the most difficult tumors to treat. The mean overall survival rate of 15 months and the 5-year survival rate of 5% have not significantly changed for almost 2 decades. Despite progress in understanding the pathophysiology of the disease, no new effective treatments to combine with radiation therapy after surgical tumor debulking have become available since the introduction of temozolomide in 1999. One of the main reasons for this is the scarcity of compounds that cross the blood-brain barrier (BBB) and reach the brain tumor tissue in therapeutically effective concentrations. In this review, we focus on the role of the BBB and its importance in developing brain tumor treatments. Moreover, we discuss drug repurposing, a drug discovery approach to identify potential effective candidates with optimal pharmacokinetic profiles for central nervous system (CNS) penetration and that allows rapid implementation in clinical trials. Additionally, we provide an overview of repurposed candidate drug currently being investigated in GBM at the preclinical and clinical levels. Finally, we highlight the importance of phase 0 trials to confirm tumor drug exposure and we discuss emerging drug delivery technologies as an alternative route to maximize therapeutic efficacy of repurposed candidate drug.
Collapse
Affiliation(s)
- Ioannis Ntafoulis
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| | - Stijn L. W. Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands;
- Department of Hospital Pharmacy, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Sieger Leenstra
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| | - Martine L. M. Lamfers
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| |
Collapse
|
14
|
Kast RE, Alfieri A, Assi HI, Burns TC, Elyamany AM, Gonzalez-Cao M, Karpel-Massler G, Marosi C, Salacz ME, Sardi I, Van Vlierberghe P, Zaghloul MS, Halatsch ME. MDACT: A New Principle of Adjunctive Cancer Treatment Using Combinations of Multiple Repurposed Drugs, with an Example Regimen. Cancers (Basel) 2022; 14:2563. [PMID: 35626167 PMCID: PMC9140192 DOI: 10.3390/cancers14102563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 12/12/2022] Open
Abstract
In part one of this two-part paper, we present eight principles that we believe must be considered for more effective treatment of the currently incurable cancers. These are addressed by multidrug adjunctive cancer treatment (MDACT), which uses multiple repurposed non-oncology drugs, not primarily to kill malignant cells, but rather to reduce the malignant cells' growth drives. Previous multidrug regimens have used MDACT principles, e.g., the CUSP9v3 glioblastoma treatment. MDACT is an amalgam of (1) the principle that to be effective in stopping a chain of events leading to an undesired outcome, one must break more than one link; (2) the principle of Palmer et al. of achieving fractional cancer cell killing via multiple drugs with independent mechanisms of action; (3) the principle of shaping versus decisive operations, both being required for successful cancer treatment; (4) an idea adapted from Chow et al., of using multiple cytotoxic medicines at low doses; (5) the idea behind CUSP9v3, using many non-oncology CNS-penetrant drugs from general medical practice, repurposed to block tumor survival paths; (6) the concept from chess that every move creates weaknesses and strengths; (7) the principle of mass-by adding force to a given effort, the chances of achieving the goal increase; and (8) the principle of blocking parallel signaling pathways. Part two gives an example MDACT regimen, gMDACT, which uses six repurposed drugs-celecoxib, dapsone, disulfiram, itraconazole, pyrimethamine, and telmisartan-to interfere with growth-driving elements common to cholangiocarcinoma, colon adenocarcinoma, glioblastoma, and non-small-cell lung cancer. gMDACT is another example of-not a replacement for-previous multidrug regimens already in clinical use, such as CUSP9v3. MDACT regimens are designed as adjuvants to be used with cytotoxic drugs.
Collapse
Affiliation(s)
| | - Alex Alfieri
- Department of Neurosurgery, Cantonal Hospital of Winterthur, 8400 Winterthur, Switzerland; (A.A.); (M.-E.H.)
| | - Hazem I. Assi
- Naef K. Basile Cancer Center, American University of Beirut, Beirut 1100, Lebanon;
| | - Terry C. Burns
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55905, USA;
| | - Ashraf M. Elyamany
- Oncology Unit, Hemato-Oncology Department, SECI Assiut University Egypt/King Saud Medical City, Riyadh 7790, Saudi Arabia;
| | - Maria Gonzalez-Cao
- Translational Cancer Research Unit, Dexeus University Hospital, 08028 Barcelona, Spain;
| | | | - Christine Marosi
- Clinical Division of Medical Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria;
| | - Michael E. Salacz
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA;
| | - Iacopo Sardi
- Department of Pediatric Oncology, Meyer Children’s Hospital, Viale Pieraccini 24, 50139 Florence, Italy;
| | - Pieter Van Vlierberghe
- Department of Biomolecular Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium;
| | - Mohamed S. Zaghloul
- Children’s Cancer Hospital & National Cancer Institute, Cairo University, Cairo 11796, Egypt;
| | - Marc-Eric Halatsch
- Department of Neurosurgery, Cantonal Hospital of Winterthur, 8400 Winterthur, Switzerland; (A.A.); (M.-E.H.)
| |
Collapse
|