Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 289
Filter
1.
Harefuah ; 162(4): 228-233, 2023 Apr.
Article in Hebrew | MEDLINE | ID: mdl-37120742

ABSTRACT

INTRODUCTION: Stereotactic radiosurgery (SRS) is a minimally invasive option commissioned in the treatment of intracranial arteriovenous malformations (AVMs). As long-term follow-up data became available, some late adverse effects have been reported, including SRS-induced neoplasia. However, the exact incidence of this adverse effect is unknown. In this article we present and discuss the topic with an unusual case of a young patient who was treated with SRS for AVM and developed a malignant brain tumor.


Subject(s)
Glioblastoma , Intracranial Arteriovenous Malformations , Radiosurgery , Humans , Follow-Up Studies , Treatment Outcome , Glioblastoma/etiology , Glioblastoma/surgery , Radiosurgery/adverse effects , Retrospective Studies , Intracranial Arteriovenous Malformations/surgery , Intracranial Arteriovenous Malformations/etiology
2.
Harefuah ; 162(4): 234-235, 2023 Apr.
Article in Hebrew | MEDLINE | ID: mdl-37120743

ABSTRACT

INTRODUCTION: Stereotactic radiosurgery is a disruptive therapeutic technique that has transformed neurosurgery and the treatment of intracranial tumors in the last few decades. Achieving tumor control rates over 90%, it is performed mostly in a single session, as an outpatient procedure involving no skin cuts, head shaving, or anesthesia, Radiosurgery stands out as a treatment modality with few and mostly transient side effects. Even though ionizing radiation (the energy used in radiosurgery) is known to be cancerogenic, radiosurgery-induced tumors have been exceedingly rare. In this issue of Harefuah, the Hadassah group reports a case of glioblastoma multiforme originating in the radio surgically treated site of an intracerebral arterio-venous malformation. We discuss what we can learn from this dire occurrence.


Subject(s)
Brain Neoplasms , Glioblastoma , Neoplasms, Radiation-Induced , Radiosurgery , Humans , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/surgery , Radiosurgery/adverse effects , Radiosurgery/methods , Glioblastoma/etiology , Glioblastoma/radiotherapy , Glioblastoma/surgery , Brain Neoplasms/etiology , Brain Neoplasms/radiotherapy , Brain Neoplasms/surgery
3.
Sci Rep ; 12(1): 12285, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35853933

ABSTRACT

Radiotherapy can induce an immunological response. One limiting factor is side effects on normal tissue. Using FLASH radiotherapy, side effects could possibly be reduced. The efficacy of FLASH in relation to conventional radiotherapy (CONV-RT) has not been extensively explored in fully immunocompetent animals. Fully immunocompetent Fischer 344 rats were inoculated with NS1 glioblastoma cells subcutaneously or intracranially. Radiotherapy was delivered with FLASH or CONV-RT at 8 Gy × 2 (subcutaneous tumors) and 12.5 Gy × 2 (intracranial tumors). Cured animals were re-challenged in order to explore long-term anti-tumor immunity. Serum analytes and gene expression were explored. The majority of animals with subcutaneous tumors were cured when treated with FLASH or CONV-RT at 8 Gy × 2. Cured animals could reject tumor re-challenge. TIMP-1 in serum was reduced in animals treated with FLASH 8 Gy × 2 compared to control animals. Animals with intracranial tumors survived longer when treated with FLASH or CONV-RT at 12.5 Gy × 2, but cure was not reached. CONV-RT and FLASH were equally effective in fully immunocompetent animals with glioblastoma. Radiotherapy was highly efficient in the subcutaneous setting, leading to cure and long-term immunity in the majority of the animals.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Brain Neoplasms/etiology , Brain Neoplasms/radiotherapy , Glioblastoma/etiology , Glioblastoma/radiotherapy , Radiotherapy/adverse effects , Radiotherapy Dosage , Rats
4.
Int J Mol Sci ; 23(2)2022 Jan 11.
Article in English | MEDLINE | ID: mdl-35054927

ABSTRACT

Background: Glioblastoma (GBM) remains a major clinical challenge due to its invasive capacity, resistance to treatment, and recurrence. We have previously shown that ODZ1 contributes to glioblastoma invasion and that ODZ1 mRNA levels can be upregulated by epigenetic mechanisms in response to hypoxia. Herein, we have further studied the transcriptional regulation of ODZ1 in GBM stem cells (GSCs) under hypoxic conditions and analyzed whether HIF2α has any role in this regulation. Methods: We performed the experiments in three primary GSC cell lines established from tumor specimens. GSCs were cultured under hypoxia, treated with HIF regulators (DMOG, chetomin), or transfected with specific siRNAs, and the expression levels of ODZ1 and HIF2α were analyzed. In addition, the response of the ODZ1 promoter cloned into a luciferase reporter plasmid to the activation of HIF was also studied. Results: The upregulation of both mRNA and protein levels of HIF2α under hypoxia conditions correlated with the expression of ODZ1 mRNA. Moreover, the knockdown of HIF2α by siRNAs downregulated the expression of ODZ1. We found, in the ODZ1 promoter, a HIF consensus binding site (GCGTG) 1358 bp from the transcription start site (TSS) and a HIF-like site (CCGTG) 826 bp from the TSS. Luciferase assays revealed that the stabilization of HIF by DMOG resulted in the increased activity of the ODZ1 promoter. Conclusions: Our data indicate that the HIF2α-mediated upregulation of ODZ1 helps strengthen the transcriptional control of this migration factor under hypoxia in glioblastoma stem cells. The discovery of this novel transcriptional pathway identifies new targets to develop strategies that may avoid GBM tumor invasion and recurrence.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/etiology , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Tenascin/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia , Cell Line, Tumor , Gene Knockdown Techniques , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/metabolism , RNA, Messenger/genetics , Tenascin/metabolism
5.
Int J Mol Sci ; 23(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35008959

ABSTRACT

Bisdemethoxycurcumin (BDMC) has biological activities, including anticancer effects in vitro; however, its anticancer effects in human glioblastoma (GBM) cells have not been examined yet. This study aimed to evaluate the tumor inhibitory effect and molecular mechanism of BDMC on human GBM 8401/luc2 cells in vitro and in vivo. In vitro studies have shown that BDMC significantly reduced cell viability and induced cell apoptosis in GBM 8401/luc2 cells. Furthermore, BDMC induced apoptosis via inhibited Bcl-2 (anti-apoptotic protein) and increased Bax (pro-apoptotic proteins) and cytochrome c release in GBM 8401/luc2 cells in vitro. Then, twelve BALB/c-nude mice were xenografted with human glioblastoma GBM 8401/luc2 cancer cells subcutaneously, and the xenograft nude mice were treated without and with BDMC (30 and 60 mg/kg of BDMC treatment) every 3 days. GBM 8401/luc2 cell xenografts experiment showed that the growth of the tumors was significantly suppressed by BDMC administration at both doses based on the reduction of tumor size and weights. BDMC did not change the body weight and the H&E histopathology analysis of liver samples, indicating that BDMC did not induce systemic toxicity. Meanwhile, treatment with BDMC up-regulated the expressions of BAX and cleaved caspase-3, while it down-regulated the protein expressions of Bcl-2 and XIAP in the tumor tissues compared with the control group. This study has demonstrated that BDMC presents potent anticancer activity on the human glioblastoma GBM 8401/luc2 cell xenograft model by inducing apoptosis and inhibiting tumor cell proliferation and shows the potential for further development to the anti-GBM cancer drug.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Diarylheptanoids/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Biobehavioral Sciences , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Gene Expression Regulation , Glioblastoma/drug therapy , Glioblastoma/etiology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Signal Transduction , Xenograft Model Antitumor Assays
6.
Semin Cancer Biol ; 86(Pt 3): 1122-1142, 2022 11.
Article in English | MEDLINE | ID: mdl-34004331

ABSTRACT

Human livelihood highly depends on applying different sources of energy whose utilization is associated with air pollution. On the other hand, air pollution may be associated with glioblastoma multiforme (GBM) development. Unlike other environmental causes of cancer (e.g., irradiation), air pollution cannot efficiently be controlled by geographical borders, regulations, and policies. The unavoidable exposure to air pollution can modify cancer incidence and mortality. GBM treatment with chemotherapy or even its surgical removal has proven insufficient (100% recurrence rate; patient's survival mean of 15 months; 90% fatality within five years) due to glioma infiltrative and migratory capacities. Given the barrage of attention and research investments currently plowed into next-generation cancer therapy, oncolytic viruses are perhaps the most vigorously pursued. Provision of an insight into the current state of the research and future direction is essential for stimulating new ideas with the potentials of filling research gaps. This review manuscript aims to overview types of brain cancer, their burden, and different causative agents. It also describes why air pollution is becoming a concerning factor. The different opinions on the association of air pollution with brain cancer are reviewed. It tries to address the significant controversy in this field by hypothesizing the air-pollution-brain-cancer association via inflammation and atopic conditions. The last section of this review deals with the oncolytic viruses, which have been used in, or are still under clinical trials for GBM treatment. Engineered adenoviruses (i.e., DNX-2401, DNX-2440, CRAd8-S-pk7 loaded Neural stem cells), herpes simplex virus type 1 (i.e., HSV-1 C134, HSV-1 rQNestin34.5v.2, HSV-1 G207, HSV-1 M032), measles virus (i.e., MV-CEA), parvovirus (i.e., ParvOryx), poliovirus (i.e., Poliovirus PVSRIPO), reovirus (i.e., pelareorep), moloney murine leukemia virus (i.e., Toca 511 vector), and vaccinia virus (i.e., vaccinia virus TG6002) as possible life-changing alleviations for GBM have been discussed. To the best of our knowledge, this review is the first review that comprehensively discusses both (i) the negative/positive association of air pollution with GBM; and (ii) the application of oncolytic viruses for GBM, including the most recent advances and clinical trials. It is also the first review that addresses the controversies over air pollution and brain cancer association. We believe that the article will significantly appeal to a broad readership of virologists, oncologists, neurologists, environmentalists, and those who work in the field of (bio)energy. Policymakers may also use it to establish better health policies and regulations about air pollution and (bio)fuels exploration, production, and consumption.


Subject(s)
Air Pollution , Brain Neoplasms , Glioblastoma , Oncolytic Virotherapy , Oncolytic Viruses , Mice , Animals , Humans , Glioblastoma/etiology , Glioblastoma/therapy , Brain Neoplasms/therapy
7.
Front Immunol ; 12: 777524, 2021.
Article in English | MEDLINE | ID: mdl-34917090

ABSTRACT

Glioblastoma (GBM) is the most common malignant primary brain tumor. Glioblastomas contain a large non-cancerous stromal compartment including various populations of tumor-associated macrophages and other myeloid cells, of which the presence was documented to correlate with malignancy and reduced survival. Via single-cell RNA sequencing of human GBM samples, only very low expression of PD-1, PD-L1 or PD-L2 could be detected, whereas the tumor micro-environment featured a marked expression of signal regulatory protein alpha (SIRPα), an inhibitory receptor present on myeloid cells, as well as its widely distributed counter-receptor CD47. CITE-Seq revealed that both SIRPα RNA and protein are prominently expressed on various populations of myeloid cells in GBM tumors, including both microglia- and monocyte-derived tumor-associated macrophages (TAMs). Similar findings were obtained in the mouse orthotopic GL261 GBM model, indicating that SIRPα is a potential target on GBM TAMs in mouse and human. A set of nanobodies, single-domain antibody fragments derived from camelid heavy chain-only antibodies, was generated against recombinant SIRPα and characterized in terms of affinity for the recombinant antigen and binding specificity on cells. Three selected nanobodies binding to mouse SIRPα were radiolabeled with 99mTc, injected in GL261 tumor-bearing mice and their biodistribution was evaluated using SPECT/CT imaging and radioactivity detection in dissected organs. Among these, Nb15 showed clear accumulation in peripheral organs such as spleen and liver, as well as a clear tumor uptake in comparison to a control non-targeting nanobody. A bivalent construct of Nb15 exhibited an increased accumulation in highly vascularized organs that express the target, such as spleen and liver, as compared to the monovalent format. However, penetration into the GL261 brain tumor fell back to levels detected with a non-targeting control nanobody. These results highlight the tumor penetration advantages of the small monovalent nanobody format and provide a qualitative proof-of-concept for using SIRPα-targeting nanobodies to noninvasively image myeloid cells in intracranial GBM tumors with high signal-to-noise ratios, even without blood-brain barrier permeabilization.


Subject(s)
Antigens, Differentiation/metabolism , Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Glioblastoma/diagnosis , Glioblastoma/metabolism , Molecular Imaging/methods , Myeloid Cells/metabolism , Receptors, Immunologic/metabolism , Single-Domain Antibodies , Animals , Antibodies, Neoplasm , Antigens, Differentiation/genetics , Biomarkers, Tumor , Brain Neoplasms/etiology , CD47 Antigen/metabolism , Cell Line, Tumor , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Glioblastoma/etiology , Host Specificity , Humans , Immunohistochemistry , Mice , Myeloid Cells/pathology , Receptors, Immunologic/genetics
8.
Sci Rep ; 11(1): 24067, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911992

ABSTRACT

Asunercept (company code APG101 [Apogenix AG]; company code CAN008 [CANbridge Pharmaceuticals]) is a novel glycosylated fusion protein that has shown promising effectiveness in glioblastoma. This Phase I study was initiated to evaluate the tolerability and safety of asunercept in combination with standard radiotherapy and temozolomide (RT/TMZ) in Asian patients with newly diagnosed glioblastoma. This was the Phase I portion of a Phase I/II open label, multicenter trial of asunercept plus standard RT/TMZ. Adults with newly-diagnosed glioblastoma received surgical resection followed by standard RT/TMZ plus asunercept 200 mg/week (Cohort 1) or 400 mg/week (Cohort 2) by 30-min IV infusion. The primary endpoint was the safety and tolerability of asunercept during concurrent asunercept and RT/TMZ; dose-limiting toxicities were observed for each dose. Secondary endpoints included pharmacokinetics (PK) and 6-month progression-free survival (PFS6). All patients (Cohort 1, n = 3; Cohort 2, n = 7) completed ≥ 7 weeks of asunercept treatment. No DLTs were experienced. Only one possibly treatment-related treatment emergent adverse event (TEAE), Grade 1 gingival swelling, was observed. No Grade > 3 TEAEs were reported and no TEAE led to treatment discontinuation. Systemic asunercept exposure increased proportionally with dose and showed low inter-patient variability. The PFS6 rate was 33.3% and 57.1% for patients in Cohort 1 and 2, respectively. Patients in Cohort 2 maintained a PFS rate of 57.1% at Month 12. Adding asunercept to standard RT/TMZ was safe and well tolerated in patients with newly-diagnosed glioblastoma and 400 mg/week resulted in encouraging efficacy.Trial registration NCT02853565, August 3, 2016.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/therapy , Glioblastoma/therapy , Radiotherapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Asian People , Biomarkers , Brain Neoplasms/diagnosis , Brain Neoplasms/etiology , Brain Neoplasms/mortality , Combined Modality Therapy , Disease Management , Drug Monitoring , Glioblastoma/diagnosis , Glioblastoma/etiology , Glioblastoma/mortality , Humans , Immunoglobulin G/administration & dosage , Kaplan-Meier Estimate , Prognosis , Recombinant Fusion Proteins/administration & dosage , Temozolomide/administration & dosage , Treatment Outcome , fas Receptor/administration & dosage
9.
Sci Rep ; 11(1): 19550, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34599235

ABSTRACT

Glioblastoma (GBM) angiogenesis is critical for tumor growth and recurrence, making it a compelling therapeutic target. Here, a disease-relevant, vascularized tumoroid in vitro model with stem-like features and stromal surrounds is reported. The model is used to recapitulate how individual components of the GBM's complex brain microenvironment such as hypoxia, vasculature-related stromal cells and growth factors support GBM angiogenesis. It is scalable, tractable, cost-effective and can be used with biologically-derived or biomimetic matrices. Patient-derived primary GBM cells are found to closely participate in blood vessel formation in contrast to a GBM cell line containing differentiated cells. Exogenous growth factors amplify this effect under normoxia but not at hypoxia suggesting that a significant amount of growth factors is already being produced under hypoxic conditions. Under hypoxia, primary GBM cells strongly co-localize with umbilical vein endothelial cells to form sprouting vascular networks, which has been reported to occur in vivo. These findings demonstrate that our 3D tumoroid in vitro model exhibits biomimetic attributes that may permit its use as a preclinical model in studying microenvironment cues of tumor angiogenesis.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Neovascularization, Pathologic , Tissue Culture Techniques , Tumor Microenvironment , Biomarkers , Brain Neoplasms/etiology , Brain Neoplasms/metabolism , Cell Line, Tumor , Fluorescent Antibody Technique , Glioblastoma/etiology , Glioblastoma/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunohistochemistry , Spheroids, Cellular
10.
J Cell Mol Med ; 25(22): 10650-10662, 2021 11.
Article in English | MEDLINE | ID: mdl-34708541

ABSTRACT

The dual-specificity tyrosine-regulated kinases DYRK1A and DYRK1B play a key role in controlling the quiescence-proliferation switch in cancer cells. Serum reduction of U87MG 2D cultures or multi-cellular tumour spheroids induced a quiescent like state characterized by increased DYRK1B and p27, and decreased pRb and cyclin D1. VER-239353 is a potent, selective inhibitor of the DYRK1A and DYRK1B kinases identified through fragment and structure-guided drug discovery. Inhibition of DYRK1A/B by VER-239353 in quiescent U87MG cells increased pRb, DYRK1B and cyclin D1 but also increased the cell cycle inhibitors p21 and p27. This resulted in exit from G0 but subsequent arrest in G1. DYRK1A/B inhibition reduced the proliferation of U87MG cells in 2D and 3D culture with greater effects observed under reduced serum conditions. Paradoxically, the induced re-expression of cell cycle proteins by DYRK1A/B inhibition further inhibited cell proliferation. Cell growth arrest induced in quiescent cells by DYRK1A/B inhibition was reversible through the addition of growth-promoting factors. DYRK inhibition-induced DNA damage and synergized with a CHK1 inhibitor in the U87MG spheroids. In vivo, DYRK1A/B inhibition-induced tumour stasis in a U87MG tumour xenograft model. These results suggest that further evaluation of VER-239353 as a treatment for glioblastoma is therefore warranted.


Subject(s)
Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Glioblastoma/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Disease Models, Animal , Female , Glioblastoma/drug therapy , Glioblastoma/etiology , Glioblastoma/pathology , Humans , Mice , Protein Kinase Inhibitors/pharmacology , Dyrk Kinases
11.
Cancer Res ; 81(23): 5935-5947, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34580063

ABSTRACT

Glioblastomas (GBM) are routinely treated with ionizing radiation (IR) but inevitably recur and develop therapy resistance. During treatment, the tissue surrounding tumors is also irradiated. IR potently induces senescence, and senescent stromal cells can promote the growth of neighboring tumor cells by secreting factors that create a senescence-associated secretory phenotype (SASP). Here, we carried out transcriptomic and tumorigenicity analyses in irradiated mouse brains to elucidate how radiotherapy-induced senescence of non-neoplastic brain cells promotes tumor growth. Following cranial irradiation, widespread senescence in the brain occurred, with the astrocytic population being particularly susceptible. Irradiated brains showed an altered transcriptomic profile characterized by upregulation of CDKN1A (p21), a key enforcer of senescence, and several SASP factors, including HGF, the ligand of the receptor tyrosine kinase (RTK) Met. Preirradiation of mouse brains increased Met-driven growth and invasiveness of orthotopically implanted glioma cells. Importantly, irradiated p21-/- mouse brains did not exhibit senescence and consequently failed to promote tumor growth. Senescent astrocytes secreted HGF to activate Met in glioma cells and to promote their migration and invasion in vitro, which could be blocked by HGF-neutralizing antibodies or the Met inhibitor crizotinib. Crizotinib also slowed the growth of glioma cells implanted in preirradiated brains. Treatment with the senolytic drug ABT-263 (navitoclax) selectively killed senescent astrocytes in vivo, significantly attenuating growth of glioma cells implanted in preirradiated brains. These results indicate that SASP factors in the irradiated tumor microenvironment drive GBM growth via RTK activation, underscoring the potential utility of adjuvant senolytic therapy for preventing GBM recurrence after radiotherapy. SIGNIFICANCE: This study uncovers mechanisms by which radiotherapy can promote GBM recurrence by inducing senescence in non-neoplastic brain cells, suggesting that senolytic therapy can blunt recurrent GBM growth and aggressiveness.


Subject(s)
Brain/pathology , Cellular Senescence , Gamma Rays/adverse effects , Glioblastoma/pathology , Neoplasm Recurrence, Local/pathology , Senescence-Associated Secretory Phenotype , Tumor Microenvironment , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain/drug effects , Brain/metabolism , Glioblastoma/drug therapy , Glioblastoma/etiology , Glioblastoma/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/etiology , Neoplasm Recurrence, Local/metabolism , Sulfonamides/pharmacology
12.
J Immunol Res ; 2021: 3412906, 2021.
Article in English | MEDLINE | ID: mdl-34557553

ABSTRACT

Glioblastoma (GBM) is the most common primary malignant brain tumor with a high mortality rate. The current treatment consists of surgical resection, radiation, and chemotherapy; however, the median survival rate is only 12-18 months despite these alternatives, highlighting the urgent need to find new strategies. The heterogeneity of GBM makes this tumor difficult to treat, and the immunotherapies result in an attractive approach to modulate the antitumoral immune responses favoring the tumor eradication. The immunotherapies for GMB including monoclonal antibodies, checkpoint inhibitors, vaccines, and oncolytic viruses, among others, have shown favorable results alone or as a multimodal treatment. In this review, we summarize and discuss promising immunotherapies for GBM currently under preclinical investigation as well as in clinical trials.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Immunotherapy , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , Brain Neoplasms/etiology , Brain Neoplasms/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , Clinical Trials as Topic , Combined Modality Therapy , Disease Management , Disease Susceptibility/immunology , Glioblastoma/etiology , Glioblastoma/metabolism , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Models, Animal , Molecular Targeted Therapy , Oncolytic Virotherapy/methods , Treatment Outcome
13.
Int J Mol Sci ; 22(12)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204510

ABSTRACT

Glioblastoma, a subset of aggressive brain tumors, deploy several means to increase blood vessel supply dedicated to the tumor mass. This includes typical program borrowed from embryonic development, such as vasculogenesis and sprouting angiogenesis, as well as unconventional processes, including co-option, vascular mimicry, and transdifferentiation, in which tumor cells are pro-actively engaged. However, these neo-generated vascular networks are morphologically and functionally abnormal, suggesting that the vascularization processes are rather inefficient in the tumor ecosystem. In this review, we reiterate the specificities of each neovascularization modality in glioblastoma, and, how they can be hampered mechanistically in the perspective of anti-cancer therapies.


Subject(s)
Glioblastoma/metabolism , Glioblastoma/pathology , Neovascularization, Pathologic/metabolism , Animals , Biomarkers , Cell Communication , Cell Differentiation , Disease Susceptibility , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Glioblastoma/etiology , Glioblastoma/therapy , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Radiation Tolerance , Signal Transduction , Tumor Microenvironment
14.
Int J Mol Sci ; 22(12)2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34207114

ABSTRACT

Haptoglobin (Hp) is a blood plasma glycoprotein that plays a critical role in tissue protection and the prevention of oxidative damage. Haptoglobin is an acute-phase protein, its concentration in plasma changes in pathology, and the test for its concentration is part of normal clinical practice. Haptoglobin is a conservative protein and is the subject of research as a potential biomarker of many diseases, including malignant neoplasms. The Human Hp gene is polymorphic and controls the synthesis of three major phenotypes-homozygous Hp1-1 and Hp2-2, and heterozygous Hp2-1, determined by a combination of allelic variants that are inherited. Numerous studies indicate that the phenotype of haptoglobin can be used to judge the individual's predisposition to various diseases. In addition, Hp undergoes various post-translational modifications (PTMs). Glioblastoma multiform (GBM) is the most malignant primary brain tumor. In our study, we have analyzed the state of Hp proteoforms in plasma and cells using 1D (SDS-PAGE) and 2D electrophoresis (2DE) with the following mass spectrometry (LC ES-MS/MS) or Western blotting. We found that the levels of α2- and ß-chain proteoforms are up-regulated in the plasma of GBM patients. An unprocessed form of Hp2-2 (PreHp2-2, zonulin) with unusual biophysical parameters (pI/Mw) was also detected in the plasma of GBM patients and glioblastoma cells. Altogether, this data shows the possibility to use proteoforms of haptoglobin as a potential GBM-specific plasma biomarker.


Subject(s)
Biomarkers, Tumor , Glioblastoma/etiology , Glioblastoma/metabolism , Haptoglobins/metabolism , Cell Line, Tumor , Chromatography, Liquid , Computational Biology/methods , Disease Susceptibility , Gene Expression Regulation, Neoplastic , Glioblastoma/diagnosis , Humans , Prognosis , Proteolysis , Proteomics/methods , Tandem Mass Spectrometry
15.
Oncogene ; 40(32): 5066-5080, 2021 08.
Article in English | MEDLINE | ID: mdl-34021259

ABSTRACT

Intratumoral heterogeneity is a characteristic of glioblastomas that contain an intermixture of cell populations displaying different glioblastoma subtype gene expression signatures. Proportions of these populations change during tumor evolution, but the occurrence and regulation of glioblastoma subtype transition is not well described. To identify regulators of glioblastoma subtypes we utilized a combination of in vitro experiments and in silico analyses, using experimentally generated as well as publicly available data. Through this combined approach SOX2 was identified to confer a proneural glioblastoma subtype gene expression signature. SFRP2 was subsequently identified as a SOX2-antagonist, able to induce a mesenchymal glioblastoma subtype signature. A subset of patient glioblastoma samples with high SFRP2 and low SOX2 expression was particularly enriched with mesenchymal subtype samples. Phenotypically, SFRP2 decreased tumor sphere formation, stemness as assessed by limiting dilution assay, and overall cell proliferation but increased cell motility, whereas SOX2 induced the opposite effects. Furthermore, an SFRP2/non-canonical-WNT/KLF4/PDGFR/phospho-AKT/SOX2 signaling axis was found to be involved in the mesenchymal transition. Analysis of human tumor tissue spatial gene expression patterns showed distinct expression of SFRP2- and SOX2-correlated genes in vascular and cellular areas, respectively. Finally, conditioned media from SFRP2 overexpressing cells increased CD206 on macrophages. Together, these findings present SFRP2 as a SOX2-antagonist with the capacity to induce a mesenchymal subtype transition in glioma cells located in vascular tumor areas, highlighting its role in glioblastoma tumor evolution and intratumoral heterogeneity.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/etiology , Glioblastoma/metabolism , Membrane Proteins/genetics , SOXB1 Transcription Factors/genetics , Carrier Proteins , Cell Line, Tumor , Gene Expression Profiling , Glioblastoma/pathology , Humans , Kruppel-Like Factor 4/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Membrane Proteins/metabolism , Organ Specificity , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction
16.
Genome Med ; 13(1): 88, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34011400

ABSTRACT

BACKGROUND: Macrophages are the most common infiltrating immune cells in gliomas and play a wide variety of pro-tumor and anti-tumor roles. However, the different subpopulations of macrophages and their effects on the tumor microenvironment remain poorly understood. METHODS: We combined new and previously published single-cell RNA-seq data from 98,015 single cells from a total of 66 gliomas to profile 19,331 individual macrophages. RESULTS: Unsupervised clustering revealed a pro-tumor subpopulation of bone marrow-derived macrophages characterized by the scavenger receptor MARCO, which is almost exclusively found in IDH1-wild-type glioblastomas. Previous studies have implicated MARCO as an unfavorable marker in melanoma and non-small cell lung cancer; here, we find that bulk MARCO expression is associated with worse prognosis and mesenchymal subtype. Furthermore, MARCO expression is significantly altered over the course of treatment with anti-PD1 checkpoint inhibitors in a response-dependent manner, which we validate with immunofluorescence imaging. CONCLUSIONS: These findings illustrate a novel macrophage subpopulation that drives tumor progression in glioblastomas and suggest potential therapeutic targets to prevent their recruitment.


Subject(s)
Biomarkers, Tumor , Glioblastoma/diagnosis , Glioblastoma/etiology , Receptors, Immunologic/genetics , Single-Cell Analysis , Tumor-Associated Macrophages/metabolism , Cell Communication/genetics , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/mortality , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Isocitrate Dehydrogenase/genetics , Mutation , Prognosis , Single-Cell Analysis/methods , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/pathology
17.
Mol Cancer Ther ; 20(7): 1305-1315, 2021 07.
Article in English | MEDLINE | ID: mdl-34001635

ABSTRACT

Glioblastoma (GBM), as the immunologically cold tumor, respond poorly to programmed cell death 1 (PD-1) immune checkpoint inhibitors because of insufficient immune infiltration. Herein, through the analysis of The Cancer Genome Atlas data and clinical glioma samples, we found Wnt/ß-catenin signal was activated in GBM and inversely related to the degree of immune cell (CD8+) infiltration and programmed cell death ligand 1 (PD-L1) expression. Blockade of Wnt/ß-catenin signal could inhibit GBM U118 cells' growth and migration, and upregulate their PD-L1 expression which indicated the possible better response to anti-PD-1 immunotherapy. Besides, in a co-culture system comprising U118 cells and Jurkat cells, Wnt inhibition alleviated Jurkat cell's apoptosis and enhanced its cytotoxic function as evidenced by obviously increased effector cytokine IFNγ secretion and lactate dehydrogenase release. Moreover, the enhanced anti-GBM effect of PD-1 antibody triggered by Wnt inhibition was observed in GL261 homograft mouse model, and the upregulation of immune cell (CD4+/CD8+) infiltration and IFNγ secretion in tumor tissues suggested that Wnt/ß-catenin inhibition could inflame cold tumor and then sensitize GBM to PD-1 blockade therapy. Taken together, our study verified the blockade of Wnt/ß-catenin signal could augment the efficacy of PD-1 blockade therapy on GBM through directly inhibiting tumor proliferation and migration, as well as facilitating T-cell infiltration and PD-L1 expression in tumor microenvironment.


Subject(s)
Glioblastoma/metabolism , Immune Checkpoint Inhibitors/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Allografts , Animals , Apoptosis , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Profiling , Glioblastoma/drug therapy , Glioblastoma/etiology , Glioblastoma/pathology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism
18.
Medicine (Baltimore) ; 100(16): e25373, 2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33879666

ABSTRACT

INTRODUCTION: Radiation induced gliomas often occurs after radiation therapy for other brain tumors. Medulloblastoma often occurs in children and its associated radiation-induced glioblastoma multiforme's (GBM) after radiotherapy often has a long latency period. Our case is very unique because the medulloblastoma was detected at an advance age and the latency period of radiation-induced GBM was relatively shorter. PATIENTS CONCERNS: A 64-year-old male was first admitted at our hospital in March 2018 with dizziness, vomiting, and blurred vision. DIAGNOSIS: Magnetic resonance imaging of brain revealed a lesion with local mixed density and mass enhancement in left cerebellar region. Histopathology established medulloblastoma (World Health Organization) grade 4 and a classic histological subtype after surgery. INTERVENTION: Surgical resection followed by radiation therapy were the initial therapeutic modalities. OUTCOMES: In April 2019, the patient was readmitted with dizziness and blurred vision. Magnetic resonance imaging showed the left cerebellar hemisphere bulky enhancement lesion. Again, a multimodal therapy comprising surgical resection, radiation therapy as well as chemotherapy was adapted after histopathology established GBM. LESION: Radiotherapy for medulloblastoma patients at advance ages is a critical predisposing factor for the development of radiation-induced GBM in a very short period of time. We suggest that, radiotherapy as adjuvant therapy for medulloblastoma patients at advance ages should be chosen with extreme caution.


Subject(s)
Cerebellar Neoplasms/therapy , Glioblastoma/etiology , Medulloblastoma/therapy , Neoplasms, Radiation-Induced/etiology , Radiotherapy, Adjuvant/adverse effects , Combined Modality Therapy , Humans , Male , Middle Aged
19.
Int J Mol Sci ; 22(8)2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33917954

ABSTRACT

Glioblastoma (GBM) is known to be the most common and lethal primary malignant brain tumor. Therapies against this neoplasia have a high percentage of failure, associated with the survival of self-renewing glioblastoma stem cells (GSCs), which repopulate treated tumors. In addition, despite new radical surgery protocols and the introduction of new anticancer drugs, protocols for treatment, and technical advances in radiotherapy, no significant improvement in the survival rate for GBMs has been realized. Thus, novel antitarget therapies could be used in conjunction with standard radiochemotherapy approaches. Targeted therapy, indeed, may address specific targets that play an essential role in the proliferation, survival, and invasiveness of GBM cells, including numerous molecules involved in signal transduction pathways. Significant cellular heterogeneity and the hierarchy with GSCs showing a therapy-resistant phenotype could explain tumor recurrence and local invasiveness and, therefore, may be a target for new therapies. Therefore, the forced differentiation of GSCs may be a promising new approach in GBM treatment. This article provides an updated review of the current standard and experimental therapies for GBM, as well as an overview of the molecular characteristics of GSCs, the mechanisms that activate resistance to current treatments, and a new antitumor strategy for treating GSCs for use as therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Drug Resistance, Neoplasm , Glioblastoma/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Animals , Antineoplastic Agents/therapeutic use , Biomarkers , Brain Neoplasms/drug therapy , Brain Neoplasms/etiology , Brain Neoplasms/pathology , Cell Differentiation , Cell Self Renewal , Disease Susceptibility , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , Glioblastoma/etiology , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects
20.
Int J Mol Sci ; 22(8)2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33918043

ABSTRACT

Glioblastoma (GBM) is the most malignant brain tumor in adults, with a dismal prognosis despite aggressive multi-modal therapy. Immunotherapy is currently being evaluated as an alternate treatment modality for recurrent GBMs in clinical trials. These immunotherapeutic approaches harness the patient's immune response to fight and eliminate tumor cells. Standard MR imaging is not adequate for response assessment to immunotherapy in GBM patients even after using refined response assessment criteria secondary to amplified immune response. Thus, there is an urgent need for the development of effective and alternative neuroimaging techniques for accurate response assessment. To this end, some groups have reported the potential of diffusion and perfusion MR imaging and amino acid-based positron emission tomography techniques in evaluating treatment response to different immunotherapeutic regimens in GBMs. The main goal of these techniques is to provide definitive metrics of treatment response at earlier time points for making informed decisions on future therapeutic interventions. This review provides an overview of available immunotherapeutic approaches used to treat GBMs. It discusses the limitations of conventional imaging and potential utilities of physiologic imaging techniques in the response assessment to immunotherapies. It also describes challenges associated with these imaging methods and potential solutions to avoid them.


Subject(s)
Brain Neoplasms/diagnostic imaging , Diagnostic Imaging , Glioblastoma/diagnostic imaging , Animals , Brain Neoplasms/etiology , Brain Neoplasms/therapy , Clinical Decision-Making , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Diagnostic Imaging/methods , Diagnostic Imaging/standards , Disease Management , Disease Susceptibility , Glioblastoma/etiology , Glioblastoma/therapy , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Magnetic Resonance Imaging , Positron-Emission Tomography , Prognosis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...