1
|
Azambuja JH, Yerneni SS, Maurer LM, Crentsil HE, Debom GN, Klei L, Smyers M, Sneiderman CT, Schwab KE, Acharya R, Wu YL, Ekambaram P, Hu D, Gough PJ, Bertin J, Melnick A, Kohanbash G, Bao R, Lucas PC, McAllister-Lucas LM. MALT1 protease inhibition restrains glioblastoma progression by reversing tumor-associated macrophage-dependent immunosuppression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.614808. [PMID: 39386586 PMCID: PMC11463364 DOI: 10.1101/2024.09.26.614808] [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
MALT1 protease is an intracellular signaling molecule that promotes tumor progression via cancer cell-intrinsic and cancer cell-extrinsic mechanisms. MALT1 has been mostly studied in lymphocytes, and little is known about its role in tumor-associated macrophages. Here, we show that MALT1 plays a key role in glioblastoma (GBM)-associated macrophages. Mechanistically, GBM tumor cells induce a MALT1-NF-κB signaling axis within macrophages, leading to macrophage migration and polarization toward an immunosuppressive phenotype. Inactivation of MALT1 protease promotes transcriptional reprogramming that reduces migration and restores a macrophage "M1-like" phenotype. Preclinical in vivo analysis shows that MALT1 inhibitor treatment results in increased immuno-reactivity of GBM-associated macrophages and reduced GBM tumor growth. Further, the addition of MALT1 inhibitor to temozolomide reduces immunosuppression in the tumor microenvironment, which may enhance the efficacy of this standard-of-care chemotherapeutic. Together, our findings suggest that MALT1 protease inhibition represents a promising macrophage-targeted immunotherapeutic strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Juliana Hofstätter Azambuja
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center; Pittsburgh, Pennsylvania
| | - Saigopalakrishna S. Yerneni
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- Department of Chemical Engineering, Carnegie Mellon University; Pittsburgh, Pennsylvania
| | - Lisa M. Maurer
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Hannah E. Crentsil
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
- Medical Scientist Training Program (MSTP), University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Gabriela N. Debom
- Department of Neurological Surgery, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Linda Klei
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Mei Smyers
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Chaim T. Sneiderman
- Department of Neurological Surgery, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Kristina E. Schwab
- Rangos Research Center Animal Imaging Core, UPMC Children’s Hospital of Pittsburgh; Pittsburgh, Pennsylvania
| | | | - Yijen Lin Wu
- Rangos Research Center Animal Imaging Core, UPMC Children’s Hospital of Pittsburgh; Pittsburgh, Pennsylvania
| | - Prasanna Ekambaram
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Dong Hu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- Department of Pathology, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Pete J. Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline; King of Prussia, Pennsylvania
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline; King of Prussia, Pennsylvania
| | - Ari Melnick
- Division of Hematology and Oncology, Cornell University, New York, New York
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Riyue Bao
- UPMC Hillman Cancer Center; Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh; Pittsburgh, Pennsylvania
| | - Peter C. Lucas
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- UPMC Hillman Cancer Center; Pittsburgh, Pennsylvania
- Department of Pathology, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
- Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Linda M. McAllister-Lucas
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center; Pittsburgh, Pennsylvania
- Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
- Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| |
Collapse
|
2
|
Altinoz MA, Yilmaz A, Taghizadehghalehjoughi A, Genc S, Yeni Y, Gecili I, Hacimuftuoglu A. Ulipristal-temozolomide-hydroxyurea combination for glioblastoma: in-vitro studies. J Neurosurg Sci 2024; 68:468-481. [PMID: 35766205 DOI: 10.23736/s0390-5616.22.05718-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a brain malignancy with worst survival. Low dose progesterone stimulates GBM growth, while progesterone receptor (PR)-antagonist mifepristone was shown to reduce growth and to enhance temozolomide sensitivity in GBM cells. Mifepristone is not available in all countries due to ethical reasons and may cause adrenal insufficiency and pelvic infections. Ulipristal is also a PR-antagonist used in treatment of uterine leiomyomas with higher biosafety. Ulipristal is demonstrated to suppress growth of breast cancer, yet it is not tested as yet whether it can also block growth and sensitize to temozolomide in glioblastoma as it was previously shown with mifepristone. Our first aim was to detect whether ulipristal exerts antiproliferative and chemotherapy-sensitizing effects in glioblastoma. Hydroxyurea inhibits DNA replication via blocking ribonucleotide reductase (RR) and it was demonstrated to increase temozolomide antineoplasticity in GBM. Progesterone receptor-activation in the uterus enhances RR transcription. Hence, we have hypothesized that PR-inactivation with ulipristal would further enhance hydroxyurea antineoplasticity by shutting down DNA synthesis mechanisms through further suppression of RR. Lastly, there exists no study as yet whether ulipristal, hydroxyurea and temozolomide could exert ternary antineoplastic efficacy, which was our last aim to define. METHODS To reveal interactions between ulipristal, hydroxyurea and temozolomide, we treated human U251 GBM cell line with these agents alone and in combination and measured cell proliferation, total antioxidant capacity (TAC) and total oxidant status (TOS) in conditioned medium and cellular cytokine gene expressions. RESULTS All agents significantly reduced cell proliferation significantly, yet the most significant decrease of GBM cells occurred with the triple drug combination at the 96th hour. All agents significantly decreased TAC and increased TOS in culture media, which was mostly relevant for the triple combination at the 96th hour. All these three agents tend to reduce the expression of immunosuppressive and/or GBM-growth stimulating cytokines TGF-β, IL-10 and IL-17 while increasing the expression of GBM-growth suppressing cytokine IL-23. CONCLUSIONS Reproposal of these agents in treatment of GBM would be a plausible approach if future studies prove their efficacy.
Collapse
Affiliation(s)
- Meric A Altinoz
- Department of Biochemistry, Acibadem University, Istanbul, Türkiye -
| | - Aysegul Yilmaz
- Department of Medical Pharmacology, Ataturk University, Erzurum, Türkiye
| | - Ali Taghizadehghalehjoughi
- Department of Veterinary Pharmacology and Toxicology, Veterinary Medicine, Ataturk University, Erzurum, Türkiye
| | - Sidika Genc
- Department of Medical Pharmacology, Ataturk University, Erzurum, Türkiye
| | - Yesim Yeni
- Department of Medical Pharmacology, Ataturk University, Erzurum, Türkiye
| | - Ibrahim Gecili
- Department of Medical Pharmacology, Ataturk University, Erzurum, Türkiye
| | | |
Collapse
|
3
|
Fisch J, de Moura AC, Feistauer V, Reinhardt LS, Molz P, Morás AM, Moura DJ, de Souza PO, Braganhol E, Almeida S, Guedes RP, Barschak AG, Giovenardi M. Effects of different maternal diets on adipose tissue inflammation and liver tissue oxidative stress in dams and their female offspring. Mol Cell Biochem 2024; 479:1257-1266. [PMID: 37354361 DOI: 10.1007/s11010-023-04791-3] [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: 01/31/2023] [Accepted: 06/15/2023] [Indexed: 06/26/2023]
Abstract
Pregnancy and lactation are important stages of fetal development. Therefore, this study investigated how different maternal diets offered during gestation and lactation periods affect adipose tissue inflammation and liver tissue oxidative stress of dams and their female offspring. Female BALB/c albino mice (60 days old) were randomized into three groups receiving a standard (CONT), hypercaloric (HD), or restricted (RD) diet during the pregnancy. After birth, female offspring weaned at 21 days were divided into two groups that received a standard or restricted diet (CONT/CONT, CONT/RD, RD/CONT, RD/RD, HD/CONT, and HD/RD) until 100 days old. Histological, oxidative parameters and inflammatory infiltrate of dams' and offspring's liver and adipose tissue were evaluated. HD dams presented non-alcoholic steatohepatitis (NASH) diagnosis and an increase in tumor necrosis factor-alpha (TNF-α) concentrations when compared to the RD and CONT dams, indicating a pro-inflammatory state. High concentrations of malondialdehyde (MDA) formation and catalase (CAT) activity in HD when compared to the CONT in the liver. SOD activity decreased in RD mice compared to CONT, and the SOD/CAT ratio was decreased in the RD and HD in comparison to the CONT. The maternal diet leads to an increase in SOD in RD/RD compared to HD/RD. RD-fed dams showed an increase in inflammatory infiltrates compared to CONT, evidencing changes caused by a restrictive diet. In the HD/CONT offspring, we verified an increase in inflammatory infiltrates in relation to the offspring fed a standard diet. In conclusion, HD, and RD, during pregnancy and lactation, altered the liver and adipose tissues of mothers. Furthermore, the maternal diet negatively impacts the offspring's adipose tissue but does not cause liver damage in these animals in adult life.
Collapse
Affiliation(s)
- Joana Fisch
- Graduate Program in Health Science, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Ana Carolina de Moura
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Vanessa Feistauer
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Luiza Steffens Reinhardt
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Patrícia Molz
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Ana Moira Morás
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Dinara Jaqueline Moura
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Priscila Oliveira de Souza
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Elizandra Braganhol
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Silvana Almeida
- Graduate Program in Health Science, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Health Science, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Alethéa Gatto Barschak
- Graduate Program in Health Science, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Márcia Giovenardi
- Graduate Program in Health Science, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil.
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil.
| |
Collapse
|
4
|
Tang F, Wang Y, Zeng Y, Xiao A, Tong A, Xu J. Tumor-associated macrophage-related strategies for glioma immunotherapy. NPJ Precis Oncol 2023; 7:78. [PMID: 37598273 PMCID: PMC10439959 DOI: 10.1038/s41698-023-00431-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/31/2023] [Indexed: 08/21/2023] Open
Abstract
High-grade glioma is one of the deadliest primary tumors of the central nervous system. Despite the many novel immunotherapies currently in development, it has been difficult to achieve breakthrough results in clinical studies. The reason may be due to the suppressive tumor microenvironment of gliomas that limits the function of specific immune cells (e.g., T cells) which are currently the primary targets of immunotherapy. However, tumor-associated macrophage, which are enriched in tumors, plays an important role in the development of GBM and is becoming a research hotspot for immunotherapy. This review focuses on current research advances in the use of macrophages as therapeutic targets or therapeutic tools for gliomas, and provides some potential research directions.
Collapse
Affiliation(s)
- Fansong Tang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuelong Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yunhui Zeng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Anqi Xiao
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
5
|
Agosti E, Panciani PP, Zeppieri M, De Maria L, Pasqualetti F, Tel A, Zanin L, Fontanella MM, Ius T. Tumor Microenvironment and Glioblastoma Cell Interplay as Promoters of Therapeutic Resistance. BIOLOGY 2023; 12:biology12050736. [PMID: 37237548 DOI: 10.3390/biology12050736] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023]
Abstract
The invasive nature of glioblastoma is problematic in a radical surgery approach and can be responsible for tumor recurrence. In order to create new therapeutic strategies, it is imperative to have a better understanding of the mechanisms behind tumor growth and invasion. The continuous cross-talk between glioma stem cells (GSCs) and the tumor microenvironment (TME) contributes to disease progression, which renders research in this field difficult and challenging. The main aim of the review was to assess the different possible mechanisms that could explain resistance to treatment promoted by TME and GSCs in glioblastoma, including the role of M2 macrophages, micro RNAs (miRNAs), and long non-coding RNAs (lncRNAs) from exosomes from the TME. A systematic review of the literature on the role of the TME in developing and promoting radioresistance and chemoresistance of GBM was performed according to PRISMA-P (Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols) guidelines. A dedicated literature review search was also performed on the immunotherapeutic agents against the immune TME. We identified 367 papers using the reported keywords. The final qualitative analysis was conducted on 25 studies. A growing amount of evidence in the current literature supports the role of M2 macrophages and non-coding RNAs in promoting the mechanisms of chemo and radioresistance. A better insight into how GBM cells interact with TME is an essential step towards comprehending the mechanisms that give rise to resistance to standard treatment, which can help to pave the way for the development of novel therapeutic strategies for GBM patients.
Collapse
Affiliation(s)
- Edoardo Agosti
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy
| | - Pier Paolo Panciani
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, Piazzale S. Maria della Misericordia 15, 33100 Udine, Italy
| | - Lucio De Maria
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy
| | - Francesco Pasqualetti
- Division of Radiation Oncology, Azienda Ospedaliero Universitaria Pisana, 56100 Pisa, Italy
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Alessandro Tel
- Clinic of Maxillofacial Surgery, Head-Neck and NeuroScience Department, University Hospital of Udine, Piazzale S. Maria della Misericordia 15, 33100 Udine, Italy
| | - Luca Zanin
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy
| | - Marco Maria Fontanella
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy
| | - Tamara Ius
- Neurosurgery Unit, Head-Neck and NeuroScience Department, University Hospital of Udine, Piazzale S. Maria della Misericordia 15, 33100 Udine, Italy
| |
Collapse
|
6
|
Smith GT, Radin DP, Tsirka SE. From protein-protein interactions to immune modulation: Therapeutic prospects of targeting Neuropilin-1 in high-grade glioma. Front Immunol 2022; 13:958620. [PMID: 36203599 PMCID: PMC9532003 DOI: 10.3389/fimmu.2022.958620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
In the past several years there has been a marked increase in our understanding of the pathophysiological hallmarks of glioblastoma development and progression, with specific respect to the contribution of the glioma tumor microenvironment to the rapid progression and treatment resistance of high-grade gliomas. Despite these strides, standard of care therapy still only targets rapidly dividing tumor cells in the glioma, and does little to curb the pro-tumorigenic functions of non-cancerous cells entrenched in the glioma microenvironment. This tumor promoting environment as well as the heterogeneity of high-grade gliomas contribute to the poor prognosis of this malignancy. The interaction of non-malignant cells in the microenvironment with the tumor cells accentuate phenotypes such as rapid proliferation or immunosuppression, so therapeutically modulating one target expressed on one cell type may be insufficient to restrain these rapidly developing neoplasias. With this in mind, identifying a target expressed on multiple cell types and understanding how it governs tumor-promoting functions in each cell type may have great utility in better managing this disease. Herein, we review the physiology and pathological effects of Neuropilin-1, a transmembrane co-receptor which mediates signal transduction pathways when associated with multiple other receptors. We discuss its effects on the properties of endothelial cells and on immune cell types within gliomas including glioma-associated macrophages, microglia, cytotoxic T cells and T regulatory cells. We also consider its effects when elaborated on the surface of tumor cells with respect to proliferation, stemness and treatment resistance, and review attempts to target Neuroplin-1 in the clinical setting.
Collapse
Affiliation(s)
- Gregory T. Smith
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Daniel P. Radin
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
- Stony Brook Medical Scientist Training Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Stella E. Tsirka
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
- Stony Brook Medical Scientist Training Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
- *Correspondence: Stella E. Tsirka,
| |
Collapse
|
7
|
Costa AK, Marqueze LFB, Gattiboni BB, Pedroso GS, Vasconcellos FF, Cunha EBB, Justa HC, Baldissera AB, Nagashima S, de Noronha L, Radak Z, Fernandes LC, Pinho RA. Physical Training Protects Against Brain Toxicity in Mice Exposed to an Experimental Model of Glioblastoma. Neurochem Res 2022; 47:3344-3354. [PMID: 35904698 DOI: 10.1007/s11064-022-03685-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/24/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022]
Abstract
Glioma 261 (Gl261) cell-mediated neurotoxicity has been reported in previous studies examining glioblastoma (GBM), and the effects of physical exercise (PE) on this neurotoxicity have been poorly investigated. This study aimed to evaluate the effects of a PE program in animals with experimental GBM. Male C57BL/6J mice were randomized into sham or GBM groups and subjected to a PE program for four weeks. Gl261 cells were administered into the intraventricular region at 48 h after the last exercise session. Body weight, water and feed consumption, and behavior were all evaluated for 21 days followed by euthanasia. The right parietal lobe was removed for the analysis of glial fibrillary acidic protein (GFAP), epidermal growth factor receptor (EGFR), vimentin, C-myc, nuclear factor kappa B (NF-κB), tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), interleukin 6 (IL-6), hydrogen peroxide, the glutathione system, and oxidative damage to proteins. The results revealed changes in the behavioral patterns of the trained animals, and no anatomopathological changes were observed in response to PE training. In contrast, animals with GBM subjected to PE exhibited lower immunoexpression of c-MYC, vimentin, and GFAP. Although experimental GBM altered the redox profile and inflammatory mediators, no significant alterations were observed after PE. In conclusion, our data provide consistent evidence of the relationship between PE and the improvement of tumorigenic parameters against the neurotoxicity of GL261 cells.
Collapse
Affiliation(s)
- Amanda K Costa
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Tech Park - Block 4, Laboratory 3. Imaculada Conceição Street, 1155, Prado Velho, Curitiba, PE, 80215-901, Brazil
| | - Luis F B Marqueze
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Tech Park - Block 4, Laboratory 3. Imaculada Conceição Street, 1155, Prado Velho, Curitiba, PE, 80215-901, Brazil
| | - Bruna B Gattiboni
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Tech Park - Block 4, Laboratory 3. Imaculada Conceição Street, 1155, Prado Velho, Curitiba, PE, 80215-901, Brazil
| | - Giulia S Pedroso
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Tech Park - Block 4, Laboratory 3. Imaculada Conceição Street, 1155, Prado Velho, Curitiba, PE, 80215-901, Brazil
| | - Franciane F Vasconcellos
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Tech Park - Block 4, Laboratory 3. Imaculada Conceição Street, 1155, Prado Velho, Curitiba, PE, 80215-901, Brazil
| | - Eduardo B B Cunha
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Tech Park - Block 4, Laboratory 3. Imaculada Conceição Street, 1155, Prado Velho, Curitiba, PE, 80215-901, Brazil
| | - Hanna C Justa
- Department of Cell Biology, Federal University of Parana, Curitiba, Brazil
| | | | - Seigo Nagashima
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Tech Park - Block 4, Laboratory 3. Imaculada Conceição Street, 1155, Prado Velho, Curitiba, PE, 80215-901, Brazil
| | - Lucia de Noronha
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Tech Park - Block 4, Laboratory 3. Imaculada Conceição Street, 1155, Prado Velho, Curitiba, PE, 80215-901, Brazil
| | - Zsolt Radak
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Luiz C Fernandes
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Ricardo A Pinho
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Tech Park - Block 4, Laboratory 3. Imaculada Conceição Street, 1155, Prado Velho, Curitiba, PE, 80215-901, Brazil.
| |
Collapse
|
8
|
Radin DP, Smith G, Moushiaveshi V, Wolf A, Bases R, Tsirka SE. Lucanthone Targets Lysosomes to Perturb Glioma Proliferation, Chemoresistance and Stemness, and Slows Tumor Growth In Vivo. Front Oncol 2022; 12:852940. [PMID: 35494072 PMCID: PMC9048484 DOI: 10.3389/fonc.2022.852940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is the most common and aggressive primary brain tumor in adults. Median survival time remains at 16-20 months despite multimodal treatment with surgical resection, radiation, temozolomide and tumor-treating fields therapy. After genotoxic stress glioma cells initiate cytoprotective autophagy, which contributes to treatment resistance, limiting the efficacy of these therapies and providing an avenue for glioma recurrence. Antagonism of autophagy steps has recently gained attention as it may enhance the efficacy of classical chemotherapies and newer immune-stimulating therapies. The modulation of autophagy in the clinic is limited by the low potency of common autophagy inhibitors and the inability of newer ones to cross the blood-brain barrier. Herein, we leverage lucanthone, an anti-schistosomal agent which crosses the blood-brain barrier and was recently reported to act as an autophagy inhibitor in breast cancer cells. Our studies show that lucanthone was toxic to glioma cells by inhibiting autophagy. It enhanced anti-glioma temozolomide (TMZ) efficacy at sub-cytotoxic concentrations, and suppressed the growth of stem-like glioma cells and temozolomide-resistant glioma stem cells. In vivo lucanthone slowed tumor growth: reduced numbers of Olig2+ glioma cells, normalized tumor vasculature, and reduced tumor hypoxia. We propose that lucanthone may serve to perturb a mechanism of temozolomide resistance and allow for successful treatment of TMZ-resistant glioblastoma.
Collapse
Affiliation(s)
- Daniel P. Radin
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
- Stony Brook Medical Scientist Training Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Gregory Smith
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Victoria Moushiaveshi
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Alexandra Wolf
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Robert Bases
- Department of Radiology, Montefiore Medical Center, New York City, NY, United States
- Department of Radiation Oncology, Montefiore Medical Center, New York City, NY, United States
| | - Stella E. Tsirka
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
- *Correspondence: Stella E. Tsirka,
| |
Collapse
|
9
|
Fabro F, Lamfers MLM, Leenstra S. Advancements, Challenges, and Future Directions in Tackling Glioblastoma Resistance to Small Kinase Inhibitors. Cancers (Basel) 2022; 14:600. [PMID: 35158868 PMCID: PMC8833415 DOI: 10.3390/cancers14030600] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Despite clinical intervention, glioblastoma (GBM) remains the deadliest brain tumor in adults. Its incurability is partly related to the establishment of drug resistance, both to standard and novel treatments. In fact, even though small kinase inhibitors have changed the standard clinical practice for several solid cancers, in GBM, they did not fulfill this promise. Drug resistance is thought to arise from the heterogeneity of GBM, which leads the development of several different mechanisms. A better understanding of the evolution and characteristics of drug resistance is of utmost importance to improve the current clinical practice. Therefore, the development of clinically relevant preclinical in vitro models which allow careful dissection of these processes is crucial to gain insights that can be translated to improved therapeutic approaches. In this review, we first discuss the heterogeneity of GBM, which is reflected in the development of several resistance mechanisms. In particular, we address the potential role of drug resistance mechanisms in the failure of small kinase inhibitors in clinical trials. Finally, we discuss strategies to overcome therapy resistance, particularly focusing on the importance of developing in vitro models, and the possible approaches that could be applied to the clinic to manage drug resistance.
Collapse
Affiliation(s)
| | | | - Sieger Leenstra
- Department of Neurosurgery, Brain Tumor Center, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (F.F.); (M.L.M.L.)
| |
Collapse
|
10
|
Krapež G, Kouter K, Jovčevska I, Videtič Paska A. Dynamic Intercell Communication between Glioblastoma and Microenvironment through Extracellular Vesicles. Biomedicines 2022; 10:biomedicines10010151. [PMID: 35052830 PMCID: PMC8773537 DOI: 10.3390/biomedicines10010151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/05/2022] [Accepted: 01/09/2022] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma is simultaneously the most common and most aggressive primary brain tumor in the central nervous system, with poor patient survival and scarce treatment options. Most primary glioblastomas reoccur and evolve radio- and chemoresistant properties which make them resistant to further treatments. Based on gene mutations and expression profiles, glioblastoma is relatively well classified; however, research shows that there is more to glioblastoma biology than that defined solely by its genetic component. Specifically, the overall malignancy of the tumor is also influenced by the dynamic communication to its immediate and distant environment, as important messengers to neighboring cells in the tumor microenvironment extracellular vesicles (EVs) have been identified. EVs and their cargo can modulate the immune microenvironment and other physiological processes, and can interact with the host immune system. They are involved in tumor cell survival and metabolism, tumor initiation, progression, and therapy resistance. However, on the other hand EVs are thought to become an effective treatment alternative, since they can cross the blood–brain barrier, are able of specific cell-targeting and can be loaded with various therapeutic molecules.
Collapse
|
11
|
Di Ianni N, Maffezzini M, Eoli M, Pellegatta S. Revisiting the Immunological Aspects of Temozolomide Considering the Genetic Landscape and the Immune Microenvironment Composition of Glioblastoma. Front Oncol 2021; 11:747690. [PMID: 34646780 PMCID: PMC8503270 DOI: 10.3389/fonc.2021.747690] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/03/2021] [Indexed: 12/13/2022] Open
Abstract
The microenvironment (ME) plays a critical role in causing glioblastoma (GBM) to be a moving and incurable target. The main features governing the interaction between cancer cells and the ME include dependency, promotion, and in rare cases, even competition. In the original Stupp protocol, the alkylating agent temozolomide (TMZ) is the first-line chemotherapy drug to treat GBM, and it is broadly used together or after radiotherapy. Some studies have described TMZ as an adjuvant to other therapeutic approaches including immunotherapy because of its ability to induce an immunogenic death of cancer cells. TMZ also exerts immunomodulatory effects on the tumor and immune ME. These findings support the coexistence of two circuits, i.e., one that subverts local immunosuppressive mechanisms and another that exerts a harmful influence on the peripheral immune response. A bias toward the latter can drive the failure of treatments based on the combination of chemotherapy and immunotherapy approaches. In this review, we will reanalyze how intrinsic and acquired resistance to TMZ impacts the immunomodulatory effects previously described by way of inducing a functional alteration of local immune cells and promoting immunosuppression and how different components of the immune ME, with particular attention to tumor-associated macrophages and microglia, can cause TMZ resistance to circumvent potential local immunogenic mechanisms.
Collapse
Affiliation(s)
- Natalia Di Ianni
- Unit of Immunotherapy of Brain Tumors, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Martina Maffezzini
- Unit of Immunotherapy of Brain Tumors, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Marica Eoli
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Serena Pellegatta
- Unit of Immunotherapy of Brain Tumors, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
12
|
Delello Di Filippo L, Hofstätter Azambuja J, Paes Dutra JA, Tavares Luiz M, Lobato Duarte J, Nicoleti LR, Olalla Saad ST, Chorilli M. Improving temozolomide biopharmaceutical properties in glioblastoma multiforme (GBM) treatment using GBM-targeting nanocarriers. Eur J Pharm Biopharm 2021; 168:76-89. [PMID: 34461214 DOI: 10.1016/j.ejpb.2021.08.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/24/2021] [Accepted: 08/22/2021] [Indexed: 12/18/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain cancer. GBM has aggressive development, and the pharmacological treatment remains a challenge due to GBM anatomical characteristics' (the blood-brain barrier and tumor microenvironment) and the increasing resistance to marketed drugs, such as temozolomide (TMZ), the first-line drug for GBM treatment. Due to physical-chemical properties such as short half-life time and the increasing resistance shown by GBM cells, high doses and repeated administrations are necessary, leading to significant adverse events. This review will discuss the main molecular mechanisms of TMZ resistance and the use of functionalized nanocarriers as an efficient and safe strategy for TMZ delivery. GBM-targeting nanocarriers are an important tool for the treatment of GBM, demonstrating to improve the biopharmaceutical properties of TMZ and repurpose its use in anti-GBM therapy. Technical aspects of nanocarriers will be discussed, and biological models highlighting the advantages and effects of functionalization strategies in TMZ anti-GBM activity. Finally, conclusions regarding the main findings will be made in the context of new perspectives for the treatment of GBM using TMZ as a chemotherapy agent, improving the sensibility and biological anti-tumor effect of TMZ through functionalization strategies.
Collapse
Affiliation(s)
| | | | | | - Marcela Tavares Luiz
- School of Pharmaceutical Science of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Jonatas Lobato Duarte
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Luiza Ribeiro Nicoleti
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Sara Teresinha Olalla Saad
- Hematology and Transfusion Medicine Center, University of Campinas (UNICAMP), Campinas 13083-970, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| |
Collapse
|
13
|
Saha D, Rabkin SD, Martuza RL. Temozolomide antagonizes oncolytic immunovirotherapy in glioblastoma. J Immunother Cancer 2021; 8:jitc-2019-000345. [PMID: 32457126 PMCID: PMC7252967 DOI: 10.1136/jitc-2019-000345] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Temozolomide (TMZ) chemotherapy is a current standard of care for glioblastoma (GBM), however it has only extended overall survival by a few months. Because it also modulates the immune system, both beneficially and negatively, understanding how TMZ interacts with immunotherapeutics is important. Oncolytic herpes simplex virus (oHSV) is a new class of cancer therapeutic with both cytotoxic and immunostimulatory activities. Here, we examine the combination of TMZ and an oHSV encoding murine interleukin 12, G47Δ-mIL12, in a mouse immunocompetent GBM model generated from non-immunogenic 005 GBM stem-like cells (GSCs. METHODS We first investigated the cytotoxic effects of TMZ and/or G47Δ-IL12 treatments in vitro, and then the antitumor effects of combination therapy in vivo in orthotopically implanted 005 GSC-derived brain tumors. To improve TMZ sensitivity, O6-methylguanine DNA methyltransferase (MGMT) was inhibited. The effects of TMZ on immune cells were evaluated by flow cytometery and immunohistochemistry. RESULTS The combination of TMZ+G47Δ-IL12 kills 005 GSCs in vitro better than single treatments. However, TMZ does not improve the survival of orthotopic tumor-bearing mice treated with G47Δ-IL12, but rather can abrogate the beneficial effects of G47Δ-IL12 when the two are given concurrently. TMZ negatively affects intratumor T cells and macrophages and splenocytes. Addition of MGMT inhibitor O6-benzylguanine (O6-BG), an inactivating pseudosubstrate of MGMT, to TMZ improved survival, but the combination with G47Δ-IL12 did not overcome the antagonistic effects of TMZ treatment on oHSV therapy. CONCLUSIONS These results illustrate that chemotherapy can adversely affect oHSV immunovirotherapy. As TMZ is the standard of care for GBM, the timing of these combined therapies should be taken into consideration when planning oHSV clinical trials with chemotherapy for GBM.
Collapse
Affiliation(s)
- Dipongkor Saha
- Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center - Abilene Campus, Abilene, Texas, USA
| | - Samuel D Rabkin
- Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Robert L Martuza
- Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Xue BZ, Xiang W, Zhang Q, Wang HF, Zhou YJ, Tian H, Abdelmaksou A, Xue J, Sun MX, Yi DY, Xiong NX, Jiang XB, Zhao HY, Fu P. CD90 low glioma-associated mesenchymal stromal/stem cells promote temozolomide resistance by activating FOXS1-mediated epithelial-mesenchymal transition in glioma cells. Stem Cell Res Ther 2021; 12:394. [PMID: 34256854 PMCID: PMC8278613 DOI: 10.1186/s13287-021-02458-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 06/14/2021] [Indexed: 12/29/2022] Open
Abstract
Background The tumour microenvironment contributes to chemotherapy resistance in gliomas, and glioma-associated mesenchymal stromal/stem cells (gaMSCs) are important stromal cell components that play multiple roles in tumour progression. However, whether gaMSCs affect chemotherapy resistance to the first-line agent temozolomide (TMZ) remains unclear. Herein, we explored the effect and mechanism of gaMSCs on resistance to TMZ in glioma cells. Methods Human glioma cells (cell line U87MG and primary glioblastoma cell line GBM-1) were cultured in conditioned media of gaMSCs and further treated with TMZ. The proliferation, apoptosis and migration of glioma cells were detected by Cell Counting Kit-8 (CCK-8), flow cytometry and wound-healing assays. The expression of FOXS1 in glioma cells was analysed by gene microarray, PCR and Western blotting. Then, FOXS1 expression in glioma cells was up- and downregulated by lentivirus transfection, and markers of the epithelial-mesenchymal transformation (EMT) process were detected. Tumour-bearing nude mice were established with different glioma cells and treated with TMZ to measure tumour size, survival time and Ki-67 expression. Finally, the expression of IL-6 in gaMSC subpopulations and its effects on FOXS1 expression in glioma cells were also investigated. Results Conditioned media of gaMSCs promoted the proliferation, migration and chemotherapy resistance of glioma cells. The increased expression of FOXS1 and activation of the EMT process in glioma cells under gaMSC-conditioned media were detected. The relationship of FOXS1, EMT and chemotherapy resistance in glioma cells was demonstrated through the regulation of FOXS1 expression in vitro and in vivo. Moreover, FOXS1 expression in glioma cells was increased by secretion of IL-6 mainly from the CD90low gaMSC subpopulation. Conclusions CD90low gaMSCs could increase FOXS1 expression in glioma cells by IL-6 secretion, thereby activating epithelial-mesenchymal transition and resistance to TMZ in glioma cells. These results indicate a new role of gaMSCs in chemotherapy resistance and provide novel therapeutic targets. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02458-8.
Collapse
Affiliation(s)
- Bing-Zhou Xue
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Xiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qing Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100050, China
| | - Hao-Fei Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu-Jie Zhou
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Han Tian
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ahmed Abdelmaksou
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Department of Neurosurgery, Faculty of Medicine, Helwan University, Cairo, 11435, Egypt
| | - Jian Xue
- Henan Vocational University of Science and Technology, Zhoukou, 466000, China
| | - Min-Xuan Sun
- Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Dong-Ye Yi
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nan-Xiang Xiong
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao-Bing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hong-Yang Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
15
|
Abstract
Extracellular vesicles (EVs) have recently garnered attention for their participation in host-microbe interactions in pneumococcal infections. However, the effect of EVs on the host immune system remain poorly understood. Our studies focus on EVs produced by Streptococcus pneumoniae (pEVs), and reveal that pEVs are internalized by macrophages, T cells, and epithelial cells. In vitro, pEVs induce NF-κB activation in a dosage-dependent manner and polarize human macrophages to an alternative (M2) phenotype. In addition, pEV pretreatment conditions macrophages to increase bacteria uptake and such macrophages may act as a reservoir for pneumococcal cells by increasing survival of the phagocytosed bacteria. When administered systemically in mice, pEVs induce cytokine release; when immobilized locally, they recruit lymphocytes and macrophages. Taken together, pEVs emerge as critical contributors to inflammatory responses and tissue damage in mammalian hosts.
Collapse
|
16
|
Combination of p38 MAPK inhibitor with PD-L1 antibody effectively prolongs survivals of temozolomide-resistant glioma-bearing mice via reduction of infiltrating glioma-associated macrophages and PD-L1 expression on resident glioma-associated microglia. Brain Tumor Pathol 2021; 38:189-200. [PMID: 34231121 DOI: 10.1007/s10014-021-00404-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/14/2021] [Indexed: 01/22/2023]
Abstract
Current conventional treatment strategies for glioblastoma (GBM) have limited efficacy due to the rapid development of resistance to temozolomide (TMZ). It is particularly urgent to develop novel therapeutic strategies that can overcome TMZ resistance and provide patients with better prognoses. Here, a TMZ-resistant GBM cell strain and a mouse model of TMZ resistance are established as valuable tools to explore novel therapeutic strategies against TMZ resistance. Experimentally, p38MAPK inhibitor reduces the accumulation of F4/80+/CD11b+ macrophages/microglia in glioma and prolongs the survivals of glioma-bearing mice. Glioma-associated macrophages/microglia have a significanct expression of PD-L1. p38MAPK inhibitor in combination with PD-L1 antibody can effectively prolongs the survivals of TMZ-resistant GBM-bearing hosts, and differentially reduces the accumulation of circulating monocytes-derived tumor-associated macrophages and PD-L1 abundances of resident glioma-associated microglia. This combination therapy could be a treatment option for patients at the recurrence or chronic TMZ maintenance stages. A clinical study to confirm the safety and effectiveness of this combination therapy is warranted.
Collapse
|
17
|
Immune System-Related Changes in Preclinical GL261 Glioblastoma under TMZ Treatment: Explaining MRSI-Based Nosological Imaging Findings with RT-PCR Analyses. Cancers (Basel) 2021; 13:cancers13112663. [PMID: 34071393 PMCID: PMC8199490 DOI: 10.3390/cancers13112663] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 01/02/2023] Open
Abstract
Glioblastomas (GB) are brain tumours with poor prognosis even after aggressive therapy. Previous work suggests that magnetic resonance spectroscopic imaging (MRSI) could act as a biomarker of efficient immune system attack onto GB, presenting oscillatory changes. Glioma-associated microglia/macrophages (GAMs) constitute the most abundant non-tumour cell type within the GB and can be polarised into anti-tumour (M1) or pro-tumour (M2) phenotypes. One of the mechanisms to mediate immunosuppression in brain tumours is the interaction between programmed cell death-1 ligand 1 (PD-L1) and programmed cell death-1 receptor (PD-1). We evaluated the subpopulations of GAMs in responding and control GB tumours to correlate PD-L1 expression to GAM polarisation in order to explain/validate MRSI-detected findings. Mice were evaluated by MRI/MRSI to assess the extent of response to treatment and with qPCR for GAMs M1 and M2 polarisation analyses. M1/M2 ratios and PD-L1 expression were higher in treated compared to control tumours. Furthermore, PD-L1 expression was positively correlated with the M1/M2 ratio. The oscillatory change in the GAMs prevailing population could be one of the key causes for the differential MRSI-detected pattern, allowing this to act as immune system activity biomarker in future work.
Collapse
|
18
|
da Cruz LLP, de Souza PO, Dal Prá M, Falchetti M, de Abreu AM, Azambuja JH, Bertoni APS, Paz AHR, Araújo AB, Visioli F, Fazolo T, da Silva GG, Worm PV, Wink MR, Zanotto-Filho A, Braganhol E. TLR4 expression and functionality are downregulated in glioblastoma cells and in tumor-associated macrophages: A new mechanism of immune evasion? Biochim Biophys Acta Mol Basis Dis 2021; 1867:166155. [PMID: 33932524 DOI: 10.1016/j.bbadis.2021.166155] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 03/30/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023]
Abstract
Glioblastoma (GB) is the most common and aggressive form of primary brain tumor, in which the presence of an inflammatory environment, composed mainly by tumor-associated macrophages (TAMs), is related to its progression and development of chemoresistance. Toll-Like Receptors (TLRs) are key components of the innate immune system and their expression in both tumor and immune-associated cells may impact the cell communication in the tumor microenvironment (TME), further modeling cancer growth and response to therapy. Here, we investigated the participation of TLR4-mediated signaling as a mechanism of induced-immune escape in GB. Initially, bioinformatics analysis of public datasets revealed that TLR4 expression is lower in GB tumors when compared to astrocytomas (AST), and in a subset of TAMs. Further, we confirmed that TLR4 expression is downregulated in chemoresistant GB, as well as in macrophages co-cultured with GB cells. Additionally, TLR4 function is impaired in those cells even following stimulation with LPS, an agonist of TLR4. Finally, experiments performed in a cohort of clinical primary and metastatic brain tumors indicated that the immunostaining of TLR4 and CD45 are inversely proportional, and confirmed the low TLR4 expression in GBs. Interestingly, the cytoplasmic/nuclear pattern of TLR4 staining in cancer tissues suggests additional roles of this receptor in carcinogenesis. Overall, our data suggest the downregulation of TLR4 expression and activity as a strategy for GB-associated immune escape. Additional studies are necessary to better understand TLR4 signaling in TME in order to improve the benefits of immunotherapy based on TLR signaling.
Collapse
Affiliation(s)
- L L P da Cruz
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - P O de Souza
- Programa de Pós-Graduação em Biociências, UFCSPA, Porto Alegre, RS, Brazil
| | - M Dal Prá
- Programa de Pós-Graduação em Biociências, UFCSPA, Porto Alegre, RS, Brazil
| | - M Falchetti
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - A M de Abreu
- Programa de Pós-Graduação em Biociências, UFCSPA, Porto Alegre, RS, Brazil
| | - J H Azambuja
- Programa de Pós-Graduação em Biociências, UFCSPA, Porto Alegre, RS, Brazil
| | - A P S Bertoni
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - A H R Paz
- Departamento de Morfologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - A B Araújo
- Centro de Processamento Celular, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil
| | - F Visioli
- Faculdade de Odontologia, UFRGS, Porto Alegre, RS, Brazil
| | - T Fazolo
- Hospital Moinhos de Vento, Porto Alegre, RS, Brazil
| | - G G da Silva
- Hospital São José, Irmandade Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Porto Alegre, RS, Brazil
| | - P V Worm
- Hospital São José, Irmandade Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Porto Alegre, RS, Brazil; Departamento de Cirurgia, UFCSPA, Porto Alegre, RS, Brazil
| | - M R Wink
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Biociências, UFCSPA, Porto Alegre, RS, Brazil
| | - A Zanotto-Filho
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - E Braganhol
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Biociências, UFCSPA, Porto Alegre, RS, Brazil; Instituto de Cardiologia do Rio Grande do Sul/Fundação Universitária do Instituto de Cardiologia (IC-FUC), Porto Alegre, RS, Brazil.
| |
Collapse
|
19
|
Tsuji S, Nakamura S, Yamada T, de Vega S, Okada Y, Inoue S, Shimazawa M, Hara H. HYBID derived from tumor cells and tumor-associated macrophages contribute to the glioblastoma growth. Brain Res 2021; 1764:147490. [PMID: 33887254 DOI: 10.1016/j.brainres.2021.147490] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/07/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022]
Abstract
Glioblastoma is the most malignant tumor of the brain associated with poor prognosis and outcome, and hence there is an urgent need to develop novel treatments for glioblastoma. In this study, we focused on hyaluronan binding protein (HYBID, as known as CEMIP/KIAA1199), a protein involved in hyaluronan depolymerization in chondrocytes and synoviocytes. We previously reported that Hybid-deficient (KO) mice show accumulation of hyaluronan in the brain, and memory impairment. To elucidate the role of HYBID in glioblastoma pathogenesis, we knocked down HYBID in human glioblastoma cells using siRNAs and developed a murine orthotopic xenograft model in the Hybid KO mice. Downregulation of HYBID in glioblastoma cells resulted in inhibition of cell proliferation and migration, and increased cell death. The growth of glioblastoma cells implanted in the mouse brain was suppressed in Hybid KO mice compared to that in the wild-type mice. Interestingly, infiltration of macrophages in the glioblastoma tissue was decreased in Hybid KO mice. Using intraperitoneal macrophages derived from Hybid KO mice and glioma cell supernatants, we examined the role of HYBID in macrophages in the tumor environment. We showed that HYBID contributes to macrophage migration and the release of pro-tumor factors. Moreover, we revealed that HYBID can be a poor prognostic factor in glioma patients by bioinformatics approaches. Our study provides data to support that HYBID expressed by both glioblastoma cells and tumor-associated macrophages may contribute to glioblastoma progression and suggests that HYBID may be a potential target for therapy that focuses on the tumor microenvironment of glioblastoma.
Collapse
Affiliation(s)
- Shohei Tsuji
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Tetsuya Yamada
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan; Department of Neurosurgery, Gifu University School of Medicine, Gifu, Japan
| | - Susana de Vega
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasunori Okada
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shintaro Inoue
- Cosmetic Health Science, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan.
| |
Collapse
|
20
|
Benecke L, Coray M, Umbricht S, Chiang D, Figueiró F, Muller L. Exosomes: Small EVs with Large Immunomodulatory Effect in Glioblastoma. Int J Mol Sci 2021; 22:3600. [PMID: 33808435 PMCID: PMC8036988 DOI: 10.3390/ijms22073600] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastomas are among the most aggressive tumors, and with low survival rates. They are characterized by the ability to create a highly immunosuppressive tumor microenvironment. Exosomes, small extracellular vesicles (EVs), mediate intercellular communication in the tumor microenvironment by transporting various biomolecules (RNA, DNA, proteins, and lipids), therefore playing a prominent role in tumor proliferation, differentiation, metastasis, and resistance to chemotherapy or radiation. Exosomes are found in all body fluids and can cross the blood-brain barrier due to their nanoscale size. Recent studies have highlighted the multiple influences of tumor-derived exosomes on immune cells. Owing to their structural and functional properties, exosomes can be an important instrument for gaining a better molecular understanding of tumors. Furthermore, they qualify not only as diagnostic and prognostic markers, but also as tools in therapies specifically targeting aggressive tumor cells, like glioblastomas.
Collapse
Affiliation(s)
- Laura Benecke
- Department of Biomedicine, University of Basel, 4051 Basel, Switzerland; (L.B.); (M.C.); (D.C.)
- Department of Otolaryngology and Head & Neck Surgery, University Hospital Basel, 4051 Basel, Switzerland
| | - Mali Coray
- Department of Biomedicine, University of Basel, 4051 Basel, Switzerland; (L.B.); (M.C.); (D.C.)
| | - Sandra Umbricht
- Faculty of Medicine, University of Basel, 4051 Basel, Switzerland;
| | - Dapi Chiang
- Department of Biomedicine, University of Basel, 4051 Basel, Switzerland; (L.B.); (M.C.); (D.C.)
| | - Fabrício Figueiró
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90035-003, Brazil;
| | - Laurent Muller
- Department of Biomedicine, University of Basel, 4051 Basel, Switzerland; (L.B.); (M.C.); (D.C.)
- Department of Otolaryngology and Head & Neck Surgery, University Hospital Basel, 4051 Basel, Switzerland
| |
Collapse
|
21
|
施 林, 李 宏, 辜 俊, 宋 憧, 李 俊, 陈 磊, 周 强, 漆 松, 陆 云. [Establishment of a mouse model bearing orthotopic temozolomide-resistant glioma]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:69-74. [PMID: 33509755 PMCID: PMC7867486 DOI: 10.12122/j.issn.1673-4254.2021.01.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To establish a mouse model bearing orthotopic temozolomide (TMZ)-resistant glioma that mimics the development of drug resistance in gliomas in vivo. METHODS Seventy-eight adult C57BL/6 mice were randomly divided into 6 groups (n=13), including 3 TMZ induced groups with low, medium and high doses (5, 25, and 50 mg/kg, respectively) and 3 control groups. In each group, 5 mice were used for evaluating tumor size, 5 for observing survival, and 3 for collecting tumor tissues for primary cell culture. In low-dose TMZ induced group, 3 mice bearing orthotopic murine glioma GL261 cell xenografts received intraperitoneal injections of 5 mg/kg TMZ for 5 days followed by a 10-day washout period before collecting glioma tissues. Tumor cell suspensions were prepared and injected in the mice in the medium-dose group, which were treated with the same protocol but with an increased TMZ dose, and the tumor cells harvested from 3 mice were injected in the high-dose group. The mice bearing GL261 cell xenografts in the 3 control groups received no treatment or were injected with medium- or high-dose TMZ. Cell colony forming assay was used to assess TMZ resistance of each generation of the tumor cells; CCK8 assay was used to determine drug resistance index of the cells. RESULTS The mouse models bearing TMZresistant glioma was successfully established. The cells from the high-dose induced group showed a significantly higher colony-forming rate than those from the high-dose control group (P < 0.05), and had a drug resistance 4.25 times higher than that of the cells from untreated control group. High-dose TMZ significantly reduced the tumor volume in the control group (P < 0.05) but not in the high-dose induced group (P < 0.01). The survival time of the tumor-bearing mice was significantly shortened in the high-dose induced group (P=0.0018). CONCLUSIONS Progressive increase of TMZ doses in mice bearing orthotopic gliomas can effectively induce TMZ resistance of the gliomas.
Collapse
Affiliation(s)
- 林勇 施
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nangfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 宏 李
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nangfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 俊伟 辜
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nangfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 憧 宋
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nangfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 俊杰 李
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nangfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 磊 陈
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nangfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 强 周
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nangfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 松涛 漆
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nangfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 云涛 陆
- />南方医科大学南方医院神经外科,广东 广州 510515Department of Neurosurgery, Nangfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
22
|
Jin K, Mao C, Chen L, Wang L, Liu Y, Yuan J. Adenosinergic Pathway: A Hope in the Immunotherapy of Glioblastoma. Cancers (Basel) 2021; 13:E229. [PMID: 33435205 PMCID: PMC7826839 DOI: 10.3390/cancers13020229] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Brain tumors comprise different types of malignancies, most of which are originated from glial cells. Glioblastoma multiforme (GBM) is the most aggressive type of brain tumor with a poor response to conventional therapies and dismal survival rates (15 months) despite multimodal therapies. The development of immunotherapeutic strategies seems to be necessary to enhance the overall survival of GBM patients. So far, the immunotherapies applied in GBM had promising results in the primary phases of clinical trials but failed to continue their beneficial effects in later phases. GBM-microenvironment (GME) is a heterogenic and rigorously immunosuppressive milieu wrapping by an impenetrable blood-brain barrier. Hence, in-depth knowledge about the dominant immunosuppressive mechanisms in the GME could foster GBM immunotherapy. Recently, the adenosinergic pathway (AP) is found to be a major player in the suppression of antitumor immune responses in the GME. Tumor cells evolve to metabolize pro-inflammatory ATP to anti-inflammatory adenosine. Adenosine can suppress immune responses through the signaling of adenosine receptors on immune cells. The preclinical results targeting AP in GBM showed promising results in reinvigorating antitumor responses, overriding chemoresistance, and increasing survival. We reviewed the current GBM immunotherapies and elaborated on the role of AP in the immunopathogenesis, treatment, and even prognosis of GBM. We suggest that future clinical studies should consider this pathway in their combination therapies along with other immunotherapeutic approaches.
Collapse
Affiliation(s)
- Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365, Renmin Eastern Road, Jinhua 321000, Zhejiang, China; (C.M.); (L.C.); (Y.L.)
| | - Chunsen Mao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365, Renmin Eastern Road, Jinhua 321000, Zhejiang, China; (C.M.); (L.C.); (Y.L.)
| | - Lin Chen
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365, Renmin Eastern Road, Jinhua 321000, Zhejiang, China; (C.M.); (L.C.); (Y.L.)
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China;
| | - Lude Wang
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China;
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365, Renmin Eastern Road, Jinhua 321000, Zhejiang, China
| | - Yuyao Liu
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365, Renmin Eastern Road, Jinhua 321000, Zhejiang, China; (C.M.); (L.C.); (Y.L.)
| | - Jianlie Yuan
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365, Renmin Eastern Road, Jinhua 321000, Zhejiang, China
| |
Collapse
|
23
|
Foray C, Valtorta S, Barca C, Winkeler A, Roll W, Müther M, Wagner S, Gardner ML, Hermann S, Schäfers M, Grauer OM, Moresco RM, Zinnhardt B, Jacobs AH. Imaging temozolomide-induced changes in the myeloid glioma microenvironment. Theranostics 2021; 11:2020-2033. [PMID: 33500706 PMCID: PMC7797694 DOI: 10.7150/thno.47269] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/21/2020] [Indexed: 12/26/2022] Open
Abstract
Rationale: The heterogeneous nature of gliomas makes the development and application of novel treatments challenging. In particular, infiltrating myeloid cells play a role in tumor progression and therapy resistance. Hence, a detailed understanding of the dynamic interplay of tumor cells and immune cells in vivo is necessary. To investigate the complex interaction between tumor progression and therapy-induced changes in the myeloid immune component of the tumor microenvironment, we used a combination of [18F]FET (amino acid metabolism) and [18F]DPA-714 (TSPO, GAMMs, tumor cells, astrocytes, endothelial cells) PET/MRI together with immune-phenotyping. The aim of the study was to monitor temozolomide (TMZ) treatment response and therapy-induced changes in the inflammatory tumor microenvironment (TME). Methods: Eighteen NMRInu/nu mice orthotopically implanted with Gli36dEGFR cells underwent MRI and PET/CT scans before and after treatment with TMZ or DMSO (vehicle). Tumor-to-background (striatum) uptake ratios were calculated and areas of unique tracer uptake (FET vs. DPA) were determined using an atlas-based volumetric approach. Results: TMZ therapy significantly modified the spatial distribution and uptake of both tracers. [18F]FET uptake was significantly reduced after therapy (-53 ± 84%) accompanied by a significant decrease of tumor volume (-17 ± 6%). In contrast, a significant increase (61 ± 33%) of [18F]DPA-714 uptake was detected by TSPO imaging in specific areas of the tumor. Immunohistochemistry (IHC) validated the reduction in tumor volumes and further revealed the presence of reactive TSPO-expressing glioma-associated microglia/macrophages (GAMMs) in the TME. Conclusion: We confirm the efficiency of [18F]FET-PET for monitoring TMZ-treatment response and demonstrate that in vivo TSPO-PET performed with [18F]DPA-714 can be used to identify specific reactive areas of myeloid cell infiltration in the TME.
Collapse
|
24
|
Wouters R, Bevers S, Riva M, De Smet F, Coosemans A. Immunocompetent Mouse Models in the Search for Effective Immunotherapy in Glioblastoma. Cancers (Basel) 2020; 13:E19. [PMID: 33374542 PMCID: PMC7793150 DOI: 10.3390/cancers13010019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive intrinsic brain tumor in adults. Despite maximal therapy consisting of surgery and radio/chemotherapy, GBM remains largely incurable with a median survival of less than 15 months. GBM has a strong immunosuppressive nature with a multitude of tumor and microenvironment (TME) derived factors that prohibit an effective immune response. To date, all clinical trials failed to provide lasting clinical efficacy, despite the relatively high success rates of preclinical studies to show effectivity of immunotherapy. Various factors may explain this discrepancy, including the inability of a single mouse model to fully recapitulate the complexity and heterogeneity of GBM. It is therefore critical to understand the features and limitations of each model, which should probably be combined to grab the full spectrum of the disease. In this review, we summarize the available knowledge concerning immune composition, stem cell characteristics and response to standard-of-care and immunotherapeutics for the most commonly available immunocompetent mouse models of GBM.
Collapse
Affiliation(s)
- Roxanne Wouters
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
- Oncoinvent, A.S., 0484 Oslo, Norway
| | - Sien Bevers
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium;
| | - Matteo Riva
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
- Department of Neurosurgery, Mont-Godinne Hospital, UCL Namur, 5530 Yvoir, Belgium
| | - Frederik De Smet
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium;
| | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
| |
Collapse
|
25
|
Molecular Characterization of Temozolomide-Treated and Non Temozolomide-Treated Glioblastoma Cells Released Extracellular Vesicles and Their Role in the Macrophage Response. Int J Mol Sci 2020; 21:ijms21218353. [PMID: 33171763 PMCID: PMC7664451 DOI: 10.3390/ijms21218353] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles (EVs) are widely investigated in glioblastoma multiforme (GBM) for their involvement in regulating GBM pathobiology as well as for their use as potential biomarkers. EVs, through cell-to-cell communication, can deliver proteins, nucleic acids, and lipids that are able to reprogram tumor-associated macrophages (TAMs). This research is aimed to concentrate, characterize, and identify molecular markers of EVs subtypes released by temozolomide (TMZ)-treated and non TMZ-treated four diverse GBM cells. Morphology, size distribution, and quantity of small (sEVs) and large (lEVs) vesicles were analyzed by cryo-TEM. Quality and quantity of EVs surface markers were evaluated, having been obtained by Western blotting. GBM cells shed a large amount of EVs, showing a cell line dependent molecular profile A comparative analysis distinguished sEVs and lEVs released by temozolomide (TMZ)-treated and non TMZ-treated GBM cells on the basis of quantity, size and markers expression. Finally, the GBM-derived sEVs and lEVs, irrespective of TMZ treatment, when challenged with macrophages, modulated cell activation toward a tendentially M2b-like phenotype.
Collapse
|
26
|
Wang B, Wang K, Jin T, Xu Q, He Y, Cui B, Wang Y. NCK1-AS1 enhances glioma cell proliferation, radioresistance and chemoresistance via miR-22-3p/IGF1R ceRNA pathway. Biomed Pharmacother 2020; 129:110395. [PMID: 32887025 DOI: 10.1016/j.biopha.2020.110395] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 06/07/2020] [Accepted: 06/13/2020] [Indexed: 12/14/2022] Open
Abstract
Glioma is the deadliest disease in human central nerve system. Abnormal expression of long noncoding RNA (lncRNA) expression has been demontrated to be implicated in various cancers. The oncogenic role of lncRNA NCK1-AS1 has been validated in cervical cancer, wheras its role in glioma remians obscure. Our research findings suggested that NCK1-AS1 was upregulated in glioma tissues and cells. NCK1-AS1 deficiency hindered cell proliferation and enhanced cell apoptosis. Additionally, the chemoresistance and radioresistance of glioma cells were impaired by NCK1-AS1 depletion. Moreover, miR-22-3p, a downstream gene of NCK1-AS1, could weaken glioma cell chemoresistance and radioresistance. Similarly, IGF1R was the downstream target gene of miR-22-3p. Further mechanism and function assays demonstrated that NCK1-AS1 promoted glioma cell growth, chemoresistance and radioresistance via sponging miR-22-3p to upregulate IGF1R. Finally, the tumor facilitator function of NCK1-AS1 was also verified by in vivo experiments. Taken together, NCK1-AS1 contributes to glioma cell proliferation, radioresistance and chemoresistance via miR-22-3p/IGF1R ceRNA pathway, which might provide a new insight for improving the radiotherapy and chemotherapy treatments of glioma.
Collapse
Affiliation(s)
- Bo Wang
- Department of Neurosurgery, Zhengzhou People's Hospital, No. 33 Huanghe Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Kai Wang
- Department of Neurosurgery, Zhengzhou People's Hospital, No. 33 Huanghe Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Tenglong Jin
- Department of Neurosurgery, Zhengzhou People's Hospital, No. 33 Huanghe Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Qiling Xu
- Department of Neurosurgery, Zhengzhou People's Hospital, No. 33 Huanghe Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Yanyang He
- Department of Neurosurgery, Zhengzhou People's Hospital, No. 33 Huanghe Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Bingzhou Cui
- Department of Neurosurgery, Zhengzhou People's Hospital, No. 33 Huanghe Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Yazhou Wang
- Department of Neurosurgery, Zhengzhou People's Hospital, No. 33 Huanghe Road, Jinshui District, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
27
|
Azambuja JH, Ludwig N, Yerneni SS, Braganhol E, Whiteside TL. Arginase-1+ Exosomes from Reprogrammed Macrophages Promote Glioblastoma Progression. Int J Mol Sci 2020; 21:E3990. [PMID: 32498400 PMCID: PMC7312363 DOI: 10.3390/ijms21113990] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 12/13/2022] Open
Abstract
Interactions between tumor cells and tumor-associated macrophages (TAMs) are critical for glioblastoma progression. The TAMs represent up to 30% of the glioblastoma mass. The role of TAMs in tumor progression and in the mechanisms underlying tumor growth remain unclear. Using an in vitro model resembling the crosstalk between macrophages and glioblastoma cells, we show that glioblastoma-derived exosomes (GBex) reprogram M1 (mediate pro-inflammatory function) and M2 (mediate anti-inflammatory function) macrophages, converting M1 into TAMs and augmenting pro-tumor functions of M2 macrophages. In turn, these GBex-reprogrammed TAMs, produce exosomes decorated by immunosuppressive and tumor-growth promoting proteins. TAM-derived exosomes disseminate these proteins in the tumor microenvironment (TME) promoting tumor cell migration and proliferation. Mechanisms underlying the promotion of glioblastoma growth involved Arginase-1+ exosomes produced by the reprogrammed TAMs. A selective Arginase-1 inhibitor, nor-NOHA reversed growth-promoting effects of Arginase-1 carried by TAM-derived exosomes. The data suggest that GBex-reprogrammed Arginase-1+ TAMs emerge as a major source of exosomes promoting tumor growth and as a potential therapeutic target in glioblastoma.
Collapse
Affiliation(s)
- Juliana H. Azambuja
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (J.H.A.); (N.L.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil;
| | - Nils Ludwig
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (J.H.A.); (N.L.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | | | - Elizandra Braganhol
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil;
| | - Theresa L. Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (J.H.A.); (N.L.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
28
|
Blockade of CD73 delays glioblastoma growth by modulating the immune environment. Cancer Immunol Immunother 2020; 69:1801-1812. [PMID: 32350590 DOI: 10.1007/s00262-020-02569-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 04/04/2020] [Indexed: 12/17/2022]
Abstract
Immunotherapy as an approach for cancer treatment is clinically promising. CD73, which is the enzyme that produces extracellular adenosine, favors cancer progression and protects the tumor from immune surveillance. While CD73 has recently been demonstrated to be a potential target for glioma treatment, its role in regulating the inflammatory tumor microenvironment has not yet been investigated. Thus, this study explores the immunotherapeutic value of the CD73 blockade in glioblastoma. The immuno-therapeutic value of the CD73 blockade was evaluated in vivo in immunocompetent pre-clinical glioblastoma model. As such, glioblastoma-bearing rats were nasally treated for 15 days with a siRNA CD73-loaded cationic-nanoemulsion (NE-siRNA CD73R). Apoptosis was determined by flow cytometry using Annexin-V staining and cell proliferation was analyzed by Ki67 expression by immunohistochemistry. The frequencies of the CD4+, CD8+, and CD4+CD25highCD39+ (Treg) T lymphocytes; CD11b+CD45high macrophages; CD11b+CD45low-microglia; and CD206+-M2-like phenotypes, along with expression levels of CD39 and CD73 in tumor and tumor-associated immune cells, were determined using flow cytometry, while inflammatory markers associated with tumor progression were evaluated using RT-qPCR. The CD73 blockade by NE-siRNA CD73 was found to induce tumor cell apoptosis. Meanwhile, the population of Tregs, microglia, and macrophages was significantly reduced in the tumor microenvironment, though IL-6, CCL17, and CCL22 increased. The treatment selectively decreased CD73 expression in the GB cells as well as in the tumor-associated-macrophages/microglia. This study indicates that CD73 knockdown using a nanotechnological approach to perform nasal delivery of siRNA-CD73 to CNS can potentially regulate the glioblastoma immune microenvironment and delay tumor growth by inducing apoptosis.
Collapse
|
29
|
Mirabdaly S, Elieh Ali Komi D, Shakiba Y, Moini A, Kiani A. Effects of temozolomide on U87MG glioblastoma cell expression of CXCR4, MMP2, MMP9, VEGF, anti-proliferatory cytotoxic and apoptotic properties. Mol Biol Rep 2020; 47:1187-1197. [PMID: 31897867 DOI: 10.1007/s11033-019-05219-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/30/2019] [Indexed: 12/13/2022]
Abstract
Temozolomide is an alkylating agent which is used in glioblastoma treatment. We aimed to investigate the effects of different concentrations of temozolomide and exposure time on U87MG glioblastoma cell expression of CXCR4, MMP2, MMP9 and VEGF. U87MG cells were cultured in different temozolomide concentrations and incubation time and the effects of temozolomide on inducing apoptosis was investigated. The levels of VEGF and CXCR4 expression were measured by RT-PCR and flowcytometry. Moreover, MMP2 and MMP9 activity and expression were assessed by ELISA and zymography. CXCR4 and VEGF expression levels decreased upon applying higher concentration of temozolomide. MMP2 and MMP-9 had lower activity in cells with longer exposure time or higher doses of temozolomide. Temozolomide induces the apoptosis in U87MG glioblastoma cells at therapeutic or higher dose. It is capable of decreasing their expression levels of VEGF and CXCR4.
Collapse
Affiliation(s)
- Seyedsaber Mirabdaly
- Students Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Shakiba
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, PO-Box: 6714869914, Kermanshah, Iran
| | - Ali Moini
- Department of Internal Medicine Imam, Reza Hospital Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Kiani
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, PO-Box: 6714869914, Kermanshah, Iran. .,Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
30
|
Azambuja JH, Ludwig N, Yerneni S, Rao A, Braganhol E, Whiteside TL. Molecular profiles and immunomodulatory activities of glioblastoma-derived exosomes. Neurooncol Adv 2020; 2:vdaa056. [PMID: 32642708 PMCID: PMC7262743 DOI: 10.1093/noajnl/vdaa056] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Glioblastoma is one of the most immunosuppressive human tumors. Emerging data suggest that glioblastoma-derived exosomes (GBex) reprogram the tumor microenvironment into a tumor-promoting milieu by mechanisms that not yet understood. METHODS Exosomes were isolated from supernatants of glioblastoma cell lines by size exclusion chromatography. The GBex endosomal origin, size, protein cargos, and ex vivo effects on immune cell functions were determined. GBex were injected intravenously into mice to evaluate their ability to in vivo modulate normal immune cell subsets. RESULTS GBex carried immunosuppressive proteins, including FasL, TRAIL, CTLA-4, CD39, and CD73, but contained few immunostimulatory proteins. GBex co-incubated with primary human immune cells induced simultaneous activation of multiple molecular pathways. In CD8+ T cells, GBex suppressed TNF-α and INF-γ release and mediated apoptosis. GBex suppressed natural killer (NK) and CD4+ T-cell activation. GBex activated the NF-κB pathway in macrophages and promoted their differentiation into M2 cells. Inhibition of the NF-κB pathway in macrophages reversed the GBex-mediated effects. GBex-driven reprogramming of macrophages involved the release of soluble factors that promoted tumor proliferation in vitro. In mice injected with GBex, the frequency of splenic CD8+ T cells, NK cells, and M1-like macrophages was reduced, while that of naïve and M2-like macrophages increased (P < .05). CONCLUSIONS GBex reprogrammed functions of all types of immune cells in vitro and altered their frequency in vivo. By creating and sustaining a highly immunosuppressive environment, GBex play a key role in promoting tumor progression.
Collapse
Affiliation(s)
- Juliana Hofstatter Azambuja
- Postgraduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Nils Ludwig
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | - Aparna Rao
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Elizandra Braganhol
- Postgraduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Theresa L Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
31
|
Teixeira FC, Bruxel F, Azambuja JH, Berenguer AM, Stefani MA, Sévigny J, Spanevello RM, Battastini AMO, Teixeira HF, Braganhol E. Development and characterization of CD73-siRNA-loaded nanoemulsion: effect on C6 glioma cells and primary astrocytes. Pharm Dev Technol 2019; 25:408-415. [DOI: 10.1080/10837450.2019.1705485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Fernanda C. Teixeira
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, UFPEL, Pelotas, Brazil
| | - Fernanda Bruxel
- Grupo de Pesquisa em Nanobiotecnologia e Nanotoxicologia, UNIPAMPA, Uruguaiana, Brazil
| | - Juliana H. Azambuja
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, UFPEL, Pelotas, Brazil
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | | | - Marco A. Stefani
- Departamento de Morfologia, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, Brazil
| | - Jean Sévigny
- Département de microbiologie-infectiologie et d’immunologie, Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec - Université Laval, Québec, Canada
| | - Roselia M. Spanevello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, UFPEL, Pelotas, Brazil
| | - Ana M. O. Battastini
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Helder F. Teixeira
- Programa de Pós-Graduação em Ciências Farmacêuticas, UFRGS, Porto Alegre, Brazil
| | - Elizandra Braganhol
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, UFPEL, Pelotas, Brazil
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| |
Collapse
|
32
|
Inhibition of the Adenosinergic Pathway in Cancer Rejuvenates Innate and Adaptive Immunity. Int J Mol Sci 2019; 20:ijms20225698. [PMID: 31739402 PMCID: PMC6888217 DOI: 10.3390/ijms20225698] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023] Open
Abstract
The adenosine pathway plays a key role in modulating immune responses in physiological and pathological conditions. Physiologically, anti-inflammatory effects of adenosine balance pro-inflammatory adenosine 5'-triphosphate (ATP), protecting tissues from damage caused by activated immune cells. Pathologically, increased adenosine monophosphatase (AMPase) activity in tumors leads to increased adenosine production, generating a deeply immunosuppressed microenvironment and promoting cancer progression. Adenosine emerges as a promising target for cancer therapy. It mediates protumor activities by inducing tumor cell proliferation, angiogenesis, chemoresistance, and migration/invasion by tumor cells. It also inhibits the functions of immune cells, promoting the formation of a tumor-permissive immune microenvironment and favoriting tumor escape from the host immune system. Pharmacologic inhibitors, siRNA or antibodies specific for the components of the adenosine pathway, or antagonists of adenosine receptors have shown efficacy in pre-clinical studies in various in vitro and in vivo tumor models and are entering the clinical arena. Inhibition of the adenosine pathway alone or in combination with classic immunotherapies offers a potentially effective therapeutic strategy in cancer.
Collapse
|
33
|
Azambuja JH, Schuh RS, Michels LR, Gelsleichter NE, Beckenkamp LR, Iser IC, Lenz GS, de Oliveira FH, Venturin G, Greggio S, daCosta JC, Wink MR, Sevigny J, Stefani MA, Battastini AMO, Teixeira HF, Braganhol E. Nasal Administration of Cationic Nanoemulsions as CD73-siRNA Delivery System for Glioblastoma Treatment: a New Therapeutical Approach. Mol Neurobiol 2019; 57:635-649. [DOI: 10.1007/s12035-019-01730-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022]
|
34
|
Valtorta S, Lo Dico A, Raccagni I, Gaglio D, Belloli S, Politi LS, Martelli C, Diceglie C, Bonanomi M, Ercoli G, Vaira V, Ottobrini L, Moresco RM. Metformin and temozolomide, a synergic option to overcome resistance in glioblastoma multiforme models. Oncotarget 2017; 8:113090-113104. [PMID: 29348889 PMCID: PMC5762574 DOI: 10.18632/oncotarget.23028] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/14/2017] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor with poor survival. Cytoreduction in association with radiotherapy and temozolomide (TMZ) is the standard therapy, but response is heterogeneous and life expectancy is limited. The combined use of chemotherapeutic agents with drugs targeting cell metabolism is becoming an interesting therapeutic option for cancer treatment. Here, we found that metformin (MET) enhances TMZ effect on TMZ-sensitive cell line (U251) and overcomes TMZ-resistance in T98G GBM cell line. In particular, combined-treatment modulated apoptosis by increasing Bax/Bcl-2 ratio, and reduced Reactive Oxygen Species (ROS) production. We also observed that MET associated with TMZ was able to reduce the expression of glioma stem cells (GSC) marker CD90 particularly in T98G cells but not that of CD133. In vivo experiments showed that combined treatment with TMZ and MET significantly slowed down growth of TMZ-resistant tumors but did not affect overall survival of TMZ-sensitive tumor bearing mice. In conclusion, our results showed that metformin is able to enhance TMZ effect in TMZ-resistant cell line suggesting its potential use in TMZ refractory GBM patients. However, the lack of effect on a GBM malignancy marker like CD133 requires further evaluation since it might influence response duration.
Collapse
Affiliation(s)
- Silvia Valtorta
- Tecnomed Foundation and Medicine and Surgery Department, University of Milan-Bicocca, Monza, Italy.,Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.,SYSBIO.IT, Centre of Systems Biology, Milan, Italy
| | - Alessia Lo Dico
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Isabella Raccagni
- Tecnomed Foundation and Medicine and Surgery Department, University of Milan-Bicocca, Monza, Italy.,Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.,SYSBIO.IT, Centre of Systems Biology, Milan, Italy
| | - Daniela Gaglio
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,SYSBIO.IT, Centre of Systems Biology, Milan, Italy
| | - Sara Belloli
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.,SYSBIO.IT, Centre of Systems Biology, Milan, Italy
| | - Letterio S Politi
- Imaging Core, IRCCS San Raffaele Scientific Institute, Milan, Italy.,University of Massachusetts Medical School, Worcester, MA, USA.,Hematology/Oncology Division and Radiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Cristina Martelli
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Cecilia Diceglie
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy.,Tecnomed Foundation and Medicine and Surgery Department, University of Milan-Bicocca, Monza, Italy
| | | | - Giulia Ercoli
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Vaira
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy.,Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luisa Ottobrini
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Rosa Maria Moresco
- Tecnomed Foundation and Medicine and Surgery Department, University of Milan-Bicocca, Monza, Italy.,Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.,SYSBIO.IT, Centre of Systems Biology, Milan, Italy
| |
Collapse
|