1
|
Cai T. Hyperbaric oxygen therapy as an adjunt treatment for glioma and brain metastasis: a literature review. Med Gas Res 2025; 15:420-426. [PMID: 39923138 PMCID: PMC12054668 DOI: 10.4103/mgr.medgasres-d-24-00096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/10/2024] [Accepted: 12/06/2024] [Indexed: 02/10/2025] Open
Abstract
The incidence and mortality rates of malignant tumors are increasing annually, with gliomas and brain metastases linked to a poor prognosis. Hyperbaric oxygen therapy is a promising treatment modality for both gliomas and brain metastases. It can alleviate tumor hypoxia and enhance radiosensitivity. When combined with other treatments for gliomas, this therapy has the potential to enhance survival rates. This review addresses the progress in research on the use of hyperbaric oxygen therapy combined with radiotherapy. For brain metastases, the combination of hyperbaric oxygen therapy and stereotactic radiosurgery is both feasible and advantagenous. This combination not only offers protection against radiation-induced brain injury but also supports the recovery of neurological and motor functions. The incidence of adverse reactions to hyperbaric oxygen therapy is relatively low, and it is safe and manageable. Future efforts should be made to investigate the mechanisms by which hyperbaric oxygen therapy combined with radiotherapy treats gliomas and brain metastases, optimize protection of the combined treatment against brain injury, minimizing adverse reactions, conducting multidisciplinary research and clinical trials, and training healthcare providers to facilitate broader clinical application.
Collapse
Affiliation(s)
- Tengteng Cai
- Department of Radiotherapy, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
2
|
Gill GS, Kharb S, Goyal G, Das P, Kurdia KC, Dhar R, Karmakar S. Immune Checkpoint Inhibitors and Immunosuppressive Tumor Microenvironment: Current Challenges and Strategies to Overcome Resistance. Immunopharmacol Immunotoxicol 2025:1-45. [PMID: 40376861 DOI: 10.1080/08923973.2025.2504906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025]
Abstract
Immune checkpoint inhibitors (ICIs) are shown to improve cancer treatment effectiveness by boosting the immune system of the patient. Nevertheless, the unique and highly suppressive TME poses a significant challenge, causing heterogeneity of response or resistance in a considerable number of patients. This review focuses on the evasive attributes of the TME. Immune evasion mechanism in TME include immunosuppressive cells, cytokine and chemokine signaling, metabolic alterations and overexpression of immune checkpoint molecules such as PD-1, CTLA-4, LAG-3, TIM-3, TIGIT, BTLA and their interactions within the TME. In addition, this review focuses on the overcoming resistance by targeting immunosuppressive cells, normalizing tumor blood vessels, blocking two or three checkpoints simultaneously, combining vaccines, oncolytic viruses and metabolic inhibitors with ICIs or other therapies. This review also focuses on the necessity of finding predictive markers for the stratification of patients and to check response of ICIs treatment. It remains to be made certain by new research and intelligent innovations how these discoveries of the TME and its interplay facilitate ICI treatment and change the face of cancer treatment.
Collapse
Affiliation(s)
- Gurpreet Singh Gill
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Simmi Kharb
- Department of Biochemistry, Pt. B.D. Sharma Postgraduate Institute of Medical Sciences, Rohtak, India
| | - Gitanjali Goyal
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kailash Chand Kurdia
- Department of GI Surgery & Liver Transplantation, All India Institute of Medical Sciences, New Delhi, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
3
|
Zhou J, Hou Z, Guan X, Zhu Z, Wang H, Wang C, Luo W, Tian C, Yang H, Ye M, Chen S, Zhang X, Zhang B. The diagnostic value of advanced tracer kinetic models in evaluating high grade gliomas recurrence and treatment response using dynamic contrast-enhanced MRI. Front Oncol 2025; 15:1536122. [PMID: 40313253 PMCID: PMC12043873 DOI: 10.3389/fonc.2025.1536122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/25/2025] [Indexed: 05/03/2025] Open
Abstract
Background The purpose of this study was to investigate the diagnostic value of advanced tracer kinetic models (TKMs) in differentiating HGGs recurrence and treatment response. Methods A total of 52 HGGs were included. DCE images were analyzed using the following TKMs: distributed parameter (DP), tissue homogeneity (TH), Brix's two-compartment (Brix) and extended-Tofts model (ETM), yielding the following parameters: cerebral blood flow (CBF), mean transit time (MTT), plasma volume (Vp), extravascular volume (Ve), vascular permeability (PS) and first-pass extraction ratio (E) in advanced TKMs (DP, TH and Brix); Ktrans, Ve, Vp and Kep in ETM. Two delineation methods were conducted (routine scans and parameter heat maps). The differences between two MRI scanners were compared. Mann-Whitney U test was used to assess the difference of parameter values. Diagnostic performance was assessed using the method of the receiver operating characteristic (ROC) curves, with the areas under the ROC curves (AUC) to determine the discriminating power of DCE parameters between recurrent tumor group and treatment response group . P<0.05 indicates statistical significance. Results The difference on the normalized kinetic parameter value (with respect to contralateral normal-appearing white matter) between two MRI scanners was statistically insignificant (P>0.05). MTT and Vp of advanced TKMs were higher in recurrent than in treatment response group (P<0.05). For ROI delineated on parameter heat maps, MTT(DP) attained the best performance with AUC 0.88, followed by MTT(TH) and Vp (DP, Brix) with AUCs around 0.80 (0.81, 0.80, 0.79 respectively). The best performance in ETM was Vp (AUC = 0.73). Conclusion MTT (DP, TH), and Vp (DP, Brix) could be potential quantitative imaging biomarkers in distinguishing recurrence and treatment response in HGGs.
Collapse
Affiliation(s)
- Jianan Zhou
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zujun Hou
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiuqi Guan
- FISCA Healthcare Co., Ltd., Nanjing, China
| | - Zhengyang Zhu
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Han Wang
- Nanjing Center for Applied Mathematics, Nanjing, China
| | - Cong Wang
- School of Electronics and Information Engineering, Suzhou Vocational University, Suzhou, China
| | - Wei Luo
- FISCA Healthcare Co., Ltd., Nanjing, China
| | - Chuanshuai Tian
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Huiquan Yang
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meiping Ye
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Sixuan Chen
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xin Zhang
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
4
|
Nateghi S, Rezayof A, Kouhkan F, Delphi L, Davisaraei YB, Rostami F, Tirgar F, Sepehri H. Growth of the prefrontal cortical glioblastoma altered cognitive and emotional behaviors via mediating miRNAs and GABA-A receptor signaling pathways in rats. Brain Res Bull 2025; 221:111227. [PMID: 39875028 DOI: 10.1016/j.brainresbull.2025.111227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025]
Abstract
The present study investigated the impact of GABAergic signaling and miRNA expression on glioblastoma multiforme (GBM) growth within the medial prefrontal cortex (mPFC) and its associated cognitive and emotional impairments. The implantation of C6 cells into the mPFC induced GBM in this brain region (referred to as the mPFC-GBM) in male Wistar rats via stereotaxic surgery, as confirmed by Magnetic Resonance Imaging (MRI), and Hematoxylin and Eosin (H&E) staining. Repeated microinjections of muscimol, a potent GABAA receptor agonist, directly into the mPFC-GBM (1 µg/rat/2.5 μl) following tumor induction decreased tumor volume and weight, resulting in an increased survival rate. Conversely, a higher dose of muscimol (6 µg/rat/2.5 μl) increased tumor size and reduced survival. Behavioral alterations induced by GBM, including anxiety-like responses, exploratory behaviors, locomotor activity, and memory formation, were assessed using anxiety-like behavior task, the hole-board test, and the novel object recognition test. Muscimol treatment dose-dependently affected these behaviors in the animals with the mPFC-GBM, bringing their performance with that of the sham group at the dose of 1 µg/rat/2.5 μl. Changes in specific miRNAs expressions, including miR-208, -290-295, -345, -743 and -802 were associated with the growth of the mPFC-GBM under muscimol treatment. These findings suggest that GBM growth into the mPFC profoundly impacts cognitive and emotional behaviors which can be improved by muscimol treatment. Considering that the expression levels of targeted miRNAs could be influenced by the growth of the mPFC-GBM, both with or without muscimol treatment, these non-coding RNAs might serve as potential biomarkers for GBM.
Collapse
Affiliation(s)
- Sepide Nateghi
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Fatemeh Kouhkan
- Stem Cell Technology Research Center (STRC), Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Ladan Delphi
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Yavar Bagheri Davisaraei
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Rostami
- Stem Cell Technology Research Center (STRC), Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Fatemeh Tirgar
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Iranian National Center for Addictions Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Houri Sepehri
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
5
|
Abdullaeva S, Chubarev V, Valeeva A, Preferanskaya N, Neganova M, Smolyarchuk E, Liu J, Sukocheva O, Samsonov M, Alyautdin R. Analysis of Clinical Success and Molecular Mechanisms of Action of Novel Anti-glioblastoma Drugs: A Review. Curr Med Chem 2025; 32:1082-1102. [PMID: 38299393 DOI: 10.2174/0109298673281283240101053940] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND Gliomas and glioblastomas (GBM) are common primary malignant brain tumors, which are highly malignant and have a poor prognosis. The presence of cancer stem cells with unrestricted proliferative capacity and ability to generate glial neoplastic cells, the diffuse nature of GBM, and other specific factors of GBM contribute to poor results of drug therapy in patients with GBM. Despite the worldwide efforts to improve the treatment, many novel anti-GBM drugs are active just in vitro, in silico, and in preclinical trials, and they sometimes demonstrate poor or no activity in clinical trials. In this paper, we have casually selected and analyzed the most promising evidence-based results related to glioblastoma treatment at FDA and Clinical Trials.gov databases. It was observed that the most prospective trend in the development of anti-GBM drugs is combination therapy vs. monotherapy. Our analysis of clinical trials has allowed us to predict that the most promising combination therapy that has shown the best results in patient's surveillance should include drugs that block different growth-promoting signals in glioblastoma cells and that are activated by the V600E BRAF mutation. One drug should inhibit signals from the BRAF protein, whereas the second drug in combination should inhibit signals from the MEK protein. METHODS The content of this review is based on information obtained from PubMed, ClinicalTrials.- gov, and the U.S. Food and Drug Administration (https://www.fda.gov/). In ClinicalTrials.gov, we retrieved studies published from January 1, 2015. In the data search, "Glioblastoma" was used as the keyword. A study was deleted if it studied remedies for concomitant tumor diseases, as well as if it did not include descriptions of treatment methods and/or if GBM was not mentioned. The analysis of the effectiveness of treatment was carried out according to the increasing overall survival in GBM patients, compared to the gold standard for this cancer. RESULTS GBM patients treated with novel immunotherapy agents and drugs acting on epigenetic factors and receptor tyrosine kinase inhibitors have shown encouraging potential for future development in clinic. However, combinations of drugs have led to more significant improvements in the results and an increase in life expectancy of patients. For example, the combination of nivolumab and ipilimumab showed a 72% increase in life expectancy compared to using nivolumab alone (9.8 vs. 16.85). CONCLUSION Combining anti-GBM drugs appears to be a key direction for increasing treatment effectiveness and overall survival. Radiotherapy of GBM can increase the effect of combination drug therapy.
Collapse
Affiliation(s)
- Sabina Abdullaeva
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
| | - Vladimir Chubarev
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
| | - Anna Valeeva
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
| | - Nina Preferanskaya
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Severnij pr. 1, Chernogolovka, 142432, Russian Federation
- Laboratory of Redox-activity Molecular System, FRC Kazan Scientific Center, Arbuzov Institute of Organic and Physical Chemistry, Russian Academy of Sciences, Akad. Arbuzov st. 8, Kazan, 420088, Russia
| | - Elena Smolyarchuk
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
| | - Junqi Liu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Olga Sukocheva
- Department of Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia
| | - Mikhail Samsonov
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
- R-Pharm JSC, Leninsky pr., 111- 1, Moscow, 119421, Russian Federation
| | - Renad Alyautdin
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
- Scientific Centre for Expert Evaluation of Medicinal Products, 8/2., Petrovsky Boulevard, Moscow, 127051, Russian Federation
| |
Collapse
|
6
|
Norollahi SE, Yousefi B, Nejatifar F, Yousefzadeh-Chabok S, Rashidy-Pour A, Samadani AA. Practical immunomodulatory landscape of glioblastoma multiforme (GBM) therapy. J Egypt Natl Canc Inst 2024; 36:33. [PMID: 39465481 DOI: 10.1186/s43046-024-00240-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 09/21/2024] [Indexed: 10/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common harmful high-grade brain tumor with high mortality and low survival rate. Importantly, besides routine diagnostic and therapeutic methods, modern and useful practical techniques are urgently needed for this serious malignancy. Correspondingly, the translational medicine focusing on genetic and epigenetic profiles of glioblastoma, as well as the immune framework and brain microenvironment, based on these challenging findings, indicates that key clinical interventions include immunotherapy, such as immunoassay, oncolytic viral therapy, and chimeric antigen receptor T (CAR T) cell therapy, which are of great importance in both diagnosis and therapy. Relatively, vaccine therapy reflects the untapped confidence to enhance GBM outcomes. Ongoing advances in immunotherapy, which utilizes different methods to regenerate or modify the resistant body for cancer therapy, have revealed serious results with many different problems and difficulties for patients. Safe checkpoint inhibitors, adoptive cellular treatment, cellular and peptide antibodies, and other innovations give researchers an endless cluster of instruments to plan profoundly in personalized medicine and the potential for combination techniques. In this way, antibodies that block immune checkpoints, particularly those that target the program death 1 (PD-1)/PD-1 (PD-L1) ligand pathway, have improved prognosis in a wide range of diseases. However, its use in combination with chemotherapy, radiation therapy, or monotherapy is ineffective in treating GBM. The purpose of this review is to provide an up-to-date overview of the translational elements concentrating on the immunotherapeutic field of GBM alongside describing the molecular mechanism involved in GBM and related signaling pathways, presenting both historical perspectives and future directions underlying basic and clinical practice.
Collapse
Affiliation(s)
- Seyedeh Elham Norollahi
- Cancer Research Center and, Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Cancer Research Center and, Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Nejatifar
- Department of Hematology and Oncology, School of Medicine, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Shahrokh Yousefzadeh-Chabok
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran
- , Rasht, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
7
|
Shen Z, Yu N, Zhang Y, Jia M, Sun Y, Li Y, Zhao L. The potential roles of HIF-1α in epithelial-mesenchymal transition and ferroptosis in tumor cells. Cell Signal 2024; 122:111345. [PMID: 39134249 DOI: 10.1016/j.cellsig.2024.111345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
In tumors, the rapid proliferation of cells and the imperfect blood supply system lead to hypoxia, which can regulate the adaptation of tumor cells to the hypoxic environment through hypoxia-inducible factor-1α (HIF-1α) and promote tumor development in multiple ways. Recent studies have found that epithelial-mesenchymal transition (EMT) and ferroptosis play important roles in the progression of tumor cells. The activation of HIF-1α is considered a key factor in inducing EMT in tumor cells. When HIF-1α is activated, it can regulate EMT-related genes, causing tumor cells to gradually lose their epithelial characteristics and acquire more invasive mesenchymal traits. The occurrence of EMT allows tumor cells to better adapt to changes in the surrounding tissue, enhancing their migratory and invasive capabilities, thus promoting tumor progression. At the same time, HIF-1α also plays a crucial regulatory role in ferroptosis in tumor cells. In a hypoxic environment, HIF-1α may affect processes such as iron metabolism and oxidative stress responses, inducing ferroptosis in tumor cells. This article briefly reviews the dual role of HIF-1α in EMT and ferroptosis in tumor cells, helping to gain a deeper understanding of the regulatory pathways of HIF-1α in the development of tumor cells, providing a new perspective for understanding the pathogenesis of tumors. The regulation of HIF-1α may become an important strategy for future tumor therapy.
Collapse
Affiliation(s)
- Zhongjun Shen
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Na Yu
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Yanfeng Zhang
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Mingbo Jia
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Ying Sun
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Yao Li
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Liyan Zhao
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China.
| |
Collapse
|
8
|
Filimonova M, Shitova A, Shevchenko L, Soldatova O, Surinova V, Rybachuk V, Kosachenko A, Nikolaev K, Volkova I, Demyashkin G, Stanojkovic TP, Zizak Z, Ivanov S, Shegay P, Kaprin A, Filimonov A. In Vitro Cytotoxic Potential and In Vivo Antitumor Effects of NOS/PDK-Inhibitor T1084. Int J Mol Sci 2024; 25:9711. [PMID: 39273658 PMCID: PMC11396549 DOI: 10.3390/ijms25179711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Previously, we showed the antitumor activity of the new NOS/PDK inhibitor T1084 (1-isobutanoyl-2-isopropylisothiourea dichloroacetate). The present study included an assessment of in vitro cytotoxicity against human malignant and normal cells according to the MTT-test and in vivo antitumor effects in solid tumor models in comparison with precursor compounds T1023 (NOS inhibitor; 1-isobutanoyl-2-isopropylisothiourea hydrobromide) and Na-DCA (PDK inhibitor; sodium dichloroacetate), using morphological, histological, and immunohistochemical methods. The effects of T1084 and T1023 on the in vitro survival of normal (MRC-5) and most malignant cells (A375, MFC-7, K562, OAW42, and PC-3) were similar and quantitatively equal. At the same time, melanoma A375 cells showed 2-2.5 times higher sensitivity (IC50: 0.39-0.41 mM) to the cytotoxicity of T1023 and T1084 than other cells. And only HeLa cells showed significantly higher sensitivity to the cytotoxicity of T1084 compared to T1023 (IC50: 0.54 ± 0.03 and 0.81 ± 0.02 mM). Comparative studies of the in vivo antitumor effects of Na-DCA, T1023, and T1084 on CC-5 cervical cancer and B-16 melanoma in mice were conducted with subchronic daily i.p. administration of these agents at an equimolar dose of 0.22 mmol/kg (33.6, 60.0, and 70.7 mg/kg, respectively). Cervical cancer CC-5 fairly quickly evaded the effects of both Na-DCA and T1023. So, from the end of the first week of Na-DCA or T1023 treatment, the tumor growth inhibition (TGI) began to decrease from 40% to an insignificant level by the end of the observation. In contrast, in two independent experiments, CC-5 showed consistently high sensitivity to the action of T1084: a significant antitumor effect with high TGI (43-58%) was registered throughout the observation, without any signs of neoplasia adaptation. The effect of precursor compounds on melanoma B-16 was either minimal (for Na-DCA) or moderate (for T1023) with TGI only 33%, which subsequently decreased by the end of the experiment. In contrast, the effect of T1084 on B-16 was qualitatively more pronounced and steadily increasing; it was accompanied by a 3-fold expansion of necrosis and dystrophy areas, a decrease in proliferation, and increased apoptosis of tumor cells. Morphologically, the T1084 effect was 2-fold superior to the effects of T1023-the TGI index reached 59-62%. This study suggests that the antitumor effects of T1084 develop through the interaction of NOS-dependent and PDK-dependent pathophysiological effects of this NOS/PDK inhibitor. The NOS inhibitory activity of T1084 exerts an anti-angiogenic effect on neoplasia. At the same time, the PDK inhibitory activity of T1084 enhances the cytotoxicity of induced intratumoral hypoxia and suppresses the development of neoplasia adaptation to anti-angiogenic stress. Such properties allow T1084 to overcome tumor resistance and realize a stable synergistic antitumor effect.
Collapse
Affiliation(s)
- Marina Filimonova
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| | - Anna Shitova
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| | - Ljudmila Shevchenko
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| | - Olga Soldatova
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| | - Valentina Surinova
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| | - Vitaly Rybachuk
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| | - Alexander Kosachenko
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| | - Kirill Nikolaev
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| | - Irina Volkova
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| | - Grigory Demyashkin
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| | - Tatjana P. Stanojkovic
- Department of Experimental Oncology, Laboratory for Radiobiology and Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Zeljko Zizak
- Department of Experimental Oncology, Laboratory for Radiobiology and Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Sergey Ivanov
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| | - Petr Shegay
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (P.S.); (A.K.)
| | - Andrey Kaprin
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (P.S.); (A.K.)
| | - Alexander Filimonov
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (L.S.); (O.S.); (V.S.); (V.R.); (A.K.); (K.N.); (I.V.); (G.D.); (A.F.)
| |
Collapse
|
9
|
Cha J, Ding EA, Carvalho EM, Fowler A, Aghi MK, Kumar S. Collagen VI deposition primes the glioblastoma microenvironment for invasion through mechanostimulation of β-catenin signaling. PNAS NEXUS 2024; 3:pgae355. [PMID: 39285933 PMCID: PMC11404513 DOI: 10.1093/pnasnexus/pgae355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024]
Abstract
While glioblastoma (GBM) progression is associated with extensive extracellular matrix (ECM) secretion, the causal contributions of ECM secretion to invasion remain unclear. Here we investigate these contributions by combining engineered materials, proteomics, analysis of patient data, and a model of bevacizumab-resistant GBM. We find that GBM cells cultured in engineered 3D hyaluronic acid hydrogels secrete ECM prior to invasion, particularly in the absence of exogenous ECM ligands. Proteomic measurements reveal extensive secretion of collagen VI, and collagen VI-associated transcripts are correspondingly enriched in microvascular proliferation regions of human GBMs. We further show that bevacizumab-resistant GBM cells deposit more collagen VI than their responsive counterparts, which is associated with marked cell-ECM stiffening. COL6A3 deletion in GBM cells reduces invasion, β-catenin signaling, and expression of mesenchymal markers, and these effects are amplified in hypoxia. Our studies strongly implicate GBM cell-derived collagen VI in microenvironmental remodeling to facilitate invasion.
Collapse
Affiliation(s)
- Junghwa Cha
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| | - Erika A Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Emily M Carvalho
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Annabelle Fowler
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| | - Manish K Aghi
- Department of Neurosurgery, University of California, San Francisco, CA 94143, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
- Department of Bioengineering and Therapeutic Sciences University of California, San Francisco, CA 94158, USA
| |
Collapse
|
10
|
Wang A, Chen P. Comprehensive analysis of circRNA-miRNA-mRNA network related to angiogenesis in recurrent implantation failure. BMC Med Genomics 2024; 17:193. [PMID: 39080700 PMCID: PMC11290139 DOI: 10.1186/s12920-024-01944-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 06/21/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Abnormal endometrial blood flow causes a decrease in endometrial receptivity and is considered a relatively independent risk factor for recurrent implantation failure (RIF). This study aimed to explore the potentially functional circRNA-miRNA-mRNA network in RIF, and further explore its mechanism. METHODS Datasets were downloaded from the GEO database to identify differentially expressed circRNAs, miRNAs and mRNAs. The circRNA-miRNA-mRNA and PPI networks were constructed using Cytoscape 3.6.0 and the STRING database, the hub genes were identified with the cytoHubba plug-in, and a circRNA-miRNA-hub mRNA regulatory sub-network was constructed. Then, GO and KEGG pathway enrichment analyses of the hub genes were performed to comprehensively analyze the mechanism of hub mRNAs in RIF. Due to the results of circRNAs-miRNAs-hub mRNAs regulatory network, we verified the expression of circRNA_0001721, circRNA_0000714, miR-17-5p, miR-29b-3p, HIF1A and VEGFA in the RIF mouse model by qRT‒PCR and western blotting. RESULTS We initially identified 175 DEmRNAs, 48 DEmiRNAs and 56 DEcircRNAs in RIF associated with angiogenesis and constructed a circRNA-miRNA‒mRNA network and PPI network. We further identified six hub genes in the acquired network. Based on these genes, functional enrichment analysis revealed that the HIF-1 signaling pathway plays a vital role in endometrial angiogenesis in RIF. In addition, the interaction networks of circRNA_0001721/miR-17-5p/HIF1A and the circRNA_0000714/miR-29b-3p/VEGFA axis were predicted. In the RIF mouse model, circRNA_0001721, circRNA_0000714, HIF1A and VEGFA were down-regulated, whereas miR-17-5p and miR-29b-3p were up-regulated according to qRT‒PCR and western blotting. CONCLUSION This study revealed that the HIF-1 signaling pathway plays a vital role in endometrial angiogenesis in RIF. The circRNA_0001721/miR-17-5p/HIF1A and circRNA_0000714/miR-29b-3p/VEGFA axes might play a role in the pathogenesis of endometrial angiogenesis in RIF.
Collapse
Affiliation(s)
- Anran Wang
- Department of Adult Chinese Medicine, Qingdao Women and Children's Hospital, Tongfu Road, Qingdao, Shandong, 266034, China.
| | - Piaopiao Chen
- Department of Orthopedics 1, Qingdao Huangdao District Second Traditional Chinese Medicine Hospital, Zhongyuan Street, Qingdao, Shandong, 266427, China
| |
Collapse
|
11
|
Li H. Melittin inactivates YAP/HIF-1α pathway via up-regulation of LATS2 to inhibit hypoxia-induced proliferation, glycolysis and angiogenesis in NSCLC. Clinics (Sao Paulo) 2024; 79:100407. [PMID: 38889502 PMCID: PMC11237868 DOI: 10.1016/j.clinsp.2024.100407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/17/2024] [Accepted: 05/13/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND NSCLC is one of the most common causes of death. The hypoxia microenvironment contributes to cancer progression. The purpose was to explore the effects and mechanism of melittin on NSCLC cells in the hypoxic microenvironment. METHODS NSCLC cell lines (A549 and H1299) were cultured in normoxia or hypoxia conditions with or without melittin treatment. The viability of the cells was detected via MTT assay and the proliferation ability was evaluated by EdU assay. QRT-PCR was performed to evaluate GLUT1, LDHA, HK2, VEGF and LATS2 mRNA levels. Glucose transport was assessed by the 2-NBDG uptake assay. The angiogenesis was determined by the tubule formation assay. The protein expressions of GLUT1, LDHA, HK2, VEGF, LATS2, YAP, p-YAP and HIF-1α were detected via western blotting assay. The tumor formation assay was conducted to examine the roles of melittin and LATS2 in vivo. RESULTS Melittin inhibited hypoxia-induced cell viability, proliferation, glycolysis and angiogenesis as well as suppressed YAP binding to HIF-1α in NSCLC. Melittin inactivated the YAP/HIF-1α pathway via up-regulation of LATS2, ultimately inhibiting cancer progression of NSCLC. Moreover, melittin suppressed tumor growth via up-regulation of LATS2 in vivo. CONCLUSION Melittin inactivated the YAP/HIF-1α pathway via up-regulation of LATS2 to contribute to the development of NSCLC. Therefore, melittin is expected to become a potential prognostic drug for the therapy of NSCLC.
Collapse
Affiliation(s)
- Hao Li
- Department of Blood Transfusion, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
12
|
Wang J, Luo T, Chen J, Liu Z, Wang J, Zhang X, Li H, Ma Y, Zhang F, Ju H, Wang W, Wang Y, Zhu Q. Enhancement of Tumor Perfusion and Antiangiogenic Therapy in Murine Models of Clear Cell Renal Cell Carcinoma Using Ultrasound-Stimulated Microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:680-689. [PMID: 38311538 DOI: 10.1016/j.ultrasmedbio.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 02/06/2024]
Abstract
OBJECTIVE To explore the effect of ultrasound-stimulated microbubble cavitation (USMC) on enhancing antiangiogenic therapy in clear cell renal cell carcinoma. MATERIALS AND METHODS We explored the effects of USMC with different mechanical indices (MIs) on tumor perfusion, 36 786-O tumor-bearing nude mice were randomly assigned into four groups: (i) control group, (ii) USMC0.25 group (MI = 0.25), (iii) USMC1.4 group (MI = 1.4) (iv) US1.4 group (MI = 1.4). Tumor perfusion was assessed by contrast-enhanced ultrasound (CEUS) before the USMC treatment and 30 min, 4h and 6h after the USMC treatment, respectively. Then we evaluated vascular normalization(VN) induced by low-MI (0.25) USMC treatment, 12 tumor-bearing nude mice were randomly divided into two groups: (i) control group (ii) USMC0.25 group. USMC treatment was performed, and tumor microvascular imaging and blood perfusion were analyzed by MicroFlow imaging (MFI) and CEUS 30 min after each treatment. In combination therapy, a total of 144 tumor-bearing nude mice were randomly assigned to six groups (n = 24): (i) control group, (ii) USMC1.4 group, (iii) USMC0.25 group, (iv) bevacizumab(BEV) group, (v) USMC1.4 +BEV group, (vi) USMC0.25 +BEV group. BEV was injected on the 6th, 10th, 14th, and 18th d after the tumors were inoculated, while USMC treatment was performed 24 h before and after every BEV administration. We examined the effects of the combination therapy through a series of experiments. RESULTS Tumor blood perfusion enhanced by USMC with low MI (0.25)could last for more than 6h, inducing tumor VN and promoting drug delivery. Compared with other groups, USMC0.25+BEV combination therapy had the strongest inhibition on tumor growth, led to the longest survival time of the mice. CONCLUSION The optimized USMC is a promising therapeutic approach that can be combined with antiangiogenic therapy to combat tumor progression.
Collapse
Affiliation(s)
- Juan Wang
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tingting Luo
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jianghong Chen
- Department of Ultrasound, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Juan Wang
- Department of Pathology,The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaolin Zhang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiiazhuang, Hebei, China
| | - Hui Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yulin Ma
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fan Zhang
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongjuan Ju
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wengang Wang
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yueheng Wang
- Department of Cardiac Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
13
|
García-Varela L, Codesido J, Perez-Pedrosa A, Muñoz-González M, Ramos-Docampo E, Rey-Bretal D, García-Otero X, Gómez-Lado N, Turrero A, Beiroa D, Rodríguez-Perez AI, Vidal A, Fernández-Ferreiro A, Pubul V, Aguiar P. Biodistribution and pharmacokinetics of [ 89Zr]-anti-VEGF mAbs using PET in glioblastoma rat models. Int J Pharm 2024; 652:123795. [PMID: 38224761 DOI: 10.1016/j.ijpharm.2024.123795] [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: 08/24/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/17/2024]
Abstract
INTRODUCTION Glioblastomas present intensive angiogenesis, thus anti-Vascular Endothelial Growth Factor (VEGF) antibodies (mAbs) have been proposed as promising therapies. However, the results of clinical trials reported moderate toxicity and limited effectiveness. This study evaluates the in vivo pharmacokinetics and biodistribution of these mAbs in a growing model of glioblastoma in rats using Positron Emission Tomography (PET). MATERIAL &Methods: mAbs were radiolabeled with zirconium-89. Four days after the model induction, animals were injected with 2.33 ± 1.3 MBq of [89Zr]-DFO-bevacizumab (n = 8) or 2.35 ± 0.26 MBq of [89Zr]-DFO-aflibercept (n = 6). PETs were performed at 0H, 48H, 168H, 240H, and 336H post-injection. Tumor induction was confirmed using [18F]-Fluorodeoxyglucose-PET and immunohistochemistry. Radiotracer uptake was estimated in all pre-defined Volumes-of-Interest. RESULTS Anti-VEGF mAbs showed 100 % Radiochemical-Purity. [89Zr]-DFO-bevacizumab showed a significantly higher bioavailability in whole-blood. A significant increase in the tumor uptake was detectable at 168H PET with [89Zr]-DFO-bevacizumab meanwhile with [89Zr]-DFO-aflibercept it was only detectable at 336H. [89Zr]-DFO-bevacizumab tumor uptake was significantly higher than that of [89Zr]-DFO-aflibercept in all the scans. Tumor induction was confirmed in all animal models. CONCLUSION MAbs detect VEGF-expression in glioblastoma models. Tumors were earlier targeted by Bevacizumab. The lower blood availability of aflibercept resulted in a lower tumor uptake than bevacizumab in all the scans.
Collapse
Affiliation(s)
- Lara García-Varela
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Jessica Codesido
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Pharmacy Dept & Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | | | - María Muñoz-González
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Emma Ramos-Docampo
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - David Rey-Bretal
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Xurxo García-Otero
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Noemí Gómez-Lado
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Angela Turrero
- Cell Cycle and Oncology Group (CiCLOn), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain
| | - Daniel Beiroa
- Centro de Biomedicina Experimental (CEBEGA), University of Santiago de Compostela, Spain
| | - Ana Isabel Rodríguez-Perez
- Cell and Molecular Neurobiology of Parkinson's disease, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain. Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Anxo Vidal
- Cell Cycle and Oncology Group (CiCLOn), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain
| | - Anxo Fernández-Ferreiro
- Pharmacy Dept & Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Virginia Pubul
- Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| | - Pablo Aguiar
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Spain
| |
Collapse
|
14
|
Cao K, Yuan W, Hou C, Wang Z, Yu J, Wang T. Hypoxic Signaling Pathways in Carotid Body Tumors. Cancers (Basel) 2024; 16:584. [PMID: 38339335 PMCID: PMC10854715 DOI: 10.3390/cancers16030584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/06/2023] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Carotid body tumors (CBTs) are rare tumors with a 1-2 incidence per 100,000 individuals. CBTs may initially present without apparent symptoms, and symptoms begin to arise since tumors grow bigger to compress surrounding tissue, such as recurrent laryngeal nerve and esophagus. Also, the etiology of CBTs remains unclear since it is more likely to occur in those who live in high-altitude areas or suffer from chronic hypoxic diseases such as COPD. SDH mutations and familial inheritance have been reported to be related to CBTs. SDH complexes play crucial roles in aerobic respiration, and SDH mutations in CBTs have been reported to be associated with hypoxia. Hypoxic signaling pathways, specifically hypoxic markers, have attracted more research attention in tumor exploration. However, the existing literature on these signaling and markers lacks a systematic review. Also, therapeutic approaches in CBTs based on hypoxic signaling are rarely used in clinics. In this review, we concluded the role of hypoxic signaling and markers and their potential implications in the initiation and progression of CBTs. Our findings underscore the involvement of the SDH family, the HIF family, VEGFs, and inflammatory cytokines (ICs) in tumorigenesis and treatment. Of particular interest is the role played by SDHx, which has recently been linked to oxygen sensing through mutations leading to hereditary CBTs. Among the SDH family, SDHB and SDHD exhibit remarkable characteristics associated with metastasis and multiple tumors. Besides SDH mutations in CBTs, the HIF family also plays crucial roles in CBTs via hypoxic signaling pathways. The HIF family regulates angiogenesis during mammalian development and tumor growth by gene expression in CBTs. HIF1α could induce the transcription of pyruvate dehydrogenase kinase 1 (PDK1) to inhibit pyruvate dehydrogenase kinase (PDH) by inhibiting the TCA cycle. Then, carotid body cells begin to hyperplasia and hypertrophy. At the same time, EPAS1 mutation, an activating mutation, could decrease the degradation of HIF2α and result in Pacak-Zhuang syndrome, which could result in paraganglioma. HIFs can also activate VEGF expression, and VEGFs act on Flk-1 to control the hyperplasia of type I cells and promote neovascularization. ICs also play a pivotal signaling role within the CB, as their expression is induced under hypoxic conditions to stimulate CB hyperplasia, ultimately leading to CBTs detecting hypoxic areas in tumors, and improving the hypoxic condition could enhance photon radiotherapy efficacy. Moreover, this review offers valuable insights for future research directions on understanding the relationship between hypoxic signaling pathways and CBTs.
Collapse
Affiliation(s)
| | | | | | | | | | - Tao Wang
- Department of Neurosurgery, Peking University Third Hospital, Beijing 100191, China; (K.C.); (W.Y.); (C.H.); (Z.W.); (J.Y.)
| |
Collapse
|
15
|
Cha J, Ding EA, Carvalho EM, Fowler A, Aghi MK, Kumar S. Glioma Cells Secrete Collagen VI to Facilitate Invasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571198. [PMID: 38168332 PMCID: PMC10760023 DOI: 10.1101/2023.12.12.571198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
While glioblastoma (GBM) progression is associated with extensive extracellular matrix (ECM) secretion, the causal contributions of ECM secretion to invasion remain unclear. Here we investigate these contributions by combining engineered materials, proteomics, analysis of patient data, and a model of bevacizumab-resistant GBM. We find that GBM cells cultured in engineered 3D hyaluronic acid hydrogels secrete ECM prior to invasion, particularly in the absence of exogenous ECM ligands. Proteomic measurements reveal extensive secretion of collagen VI, and collagen VI-associated transcripts are correspondingly enriched in microvascular proliferation regions of human GBMs. We further show that bevacizumab-resistant GBM cells deposit more collagen VI than their responsive counterparts, which is associated with marked cell-ECM stiffening. COL6A3 deletion in GBM cells reduces invasion, β-catenin signaling, and expression of mesenchymal markers, and these effects are amplified in hypoxia. Our studies strongly implicate GBM cell-derived collagen VI in microenvironmental remodeling to facilitate invasion.
Collapse
Affiliation(s)
- Junghwa Cha
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| | - Erika A Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Emily M Carvalho
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Annabelle Fowler
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| | - Manish K Aghi
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
- Department of Bioengineering and Therapeutic Sciences University of California San Francisco, CA 94158, USA
| |
Collapse
|
16
|
Abou Khouzam R, Janji B, Thiery J, Zaarour RF, Chamseddine AN, Mayr H, Savagner P, Kieda C, Gad S, Buart S, Lehn JM, Limani P, Chouaib S. Hypoxia as a potential inducer of immune tolerance, tumor plasticity and a driver of tumor mutational burden: Impact on cancer immunotherapy. Semin Cancer Biol 2023; 97:104-123. [PMID: 38029865 DOI: 10.1016/j.semcancer.2023.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023]
Abstract
In cancer patients, immune cells are often functionally compromised due to the immunosuppressive features of the tumor microenvironment (TME) which contribute to the failures in cancer therapies. Clinical and experimental evidence indicates that developing tumors adapt to the immunological environment and create a local microenvironment that impairs immune function by inducing immune tolerance and invasion. In this context, microenvironmental hypoxia, which is an established hallmark of solid tumors, significantly contributes to tumor aggressiveness and therapy resistance through the induction of tumor plasticity/heterogeneity and, more importantly, through the differentiation and expansion of immune-suppressive stromal cells. We and others have provided evidence indicating that hypoxia also drives genomic instability in cancer cells and interferes with DNA damage response and repair suggesting that hypoxia could be a potential driver of tumor mutational burden. Here, we reviewed the current knowledge on how hypoxic stress in the TME impacts tumor angiogenesis, heterogeneity, plasticity, and immune resistance, with a special interest in tumor immunogenicity and hypoxia targeting. An integrated understanding of the complexity of the effect of hypoxia on the immune and microenvironmental components could lead to the identification of better adapted and more effective combinational strategies in cancer immunotherapy. Clearly, the discovery and validation of therapeutic targets derived from the hypoxic tumor microenvironment is of major importance and the identification of critical hypoxia-associated pathways could generate targets that are undeniably attractive for combined cancer immunotherapy approaches.
Collapse
Affiliation(s)
- Raefa Abou Khouzam
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates.
| | - Bassam Janji
- Department of Cancer Research, Luxembourg Institute of Health, Tumor Immunotherapy and Microenvironment (TIME) Group, 6A, rue Nicolas-Ernest Barblé, L-1210 Luxembourg city, Luxembourg.
| | - Jerome Thiery
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculty of Medicine, University Paris-Saclay, 94805 Villejuif, France.
| | - Rania Faouzi Zaarour
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates.
| | - Ali N Chamseddine
- Gastroenterology Department, Cochin University Hospital, Université de Paris, APHP, Paris, France; Ambroise Paré - Hartmann Private Hospital Group, Oncology Unit, Neuilly-sur-Seine, France.
| | - Hemma Mayr
- Swiss Hepato-Pancreato-Biliary (HPB) and Transplantation Center, University Hospital Zurich, Raemistrasse 100, Zurich, Switzerland; Department of Surgery & Transplantation, University and University Hospital Zurich, Raemistrasse 100, Zurich, Switzerland.
| | - Pierre Savagner
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculty of Medicine, University Paris-Saclay, 94805 Villejuif, France.
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; Centre for Molecular Biophysics, UPR 4301 CNRS, 45071 Orleans, France; Centre of Postgraduate Medical Education, 01-004 Warsaw, Poland.
| | - Sophie Gad
- Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences Lettres University (PSL), 75014 Paris, France; UMR CNRS 9019, Genome Integrity and Cancers, Gustave Roussy, Paris-Saclay University, 94800 Villejuif, France.
| | - Stéphanie Buart
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculty of Medicine, University Paris-Saclay, 94805 Villejuif, France.
| | - Jean-Marie Lehn
- Institut de Science et d'Ingénierie Supramoléculaires (ISIS), Université de Strasbourg, 8 allée Gaspard Monge, Strasbourg, France.
| | - Perparim Limani
- Swiss Hepato-Pancreato-Biliary (HPB) and Transplantation Center, University Hospital Zurich, Raemistrasse 100, Zurich, Switzerland; Department of Surgery & Transplantation, University and University Hospital Zurich, Raemistrasse 100, Zurich, Switzerland.
| | - Salem Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates; INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculty of Medicine, University Paris-Saclay, 94805 Villejuif, France.
| |
Collapse
|
17
|
Blanchard R, Adjei I. Engineering the glioblastoma microenvironment with bioactive nanoparticles for effective immunotherapy. RSC Adv 2023; 13:31411-31425. [PMID: 37901257 PMCID: PMC10603567 DOI: 10.1039/d3ra01153d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
While immunotherapies have revolutionized treatment for other cancers, glioblastoma multiforme (GBM) patients have not shown similar positive responses. The limited response to immunotherapies is partly due to the unique challenges associated with the GBM tumor microenvironment (TME), which promotes resistance to immunotherapies, causing many promising therapies to fail. There is, therefore, an urgent need to develop strategies that make the TME immune permissive to promote treatment efficacy. Bioactive nano-delivery systems, in which the nanoparticle, due to its chemical composition, provides the pharmacological function, have recently emerged as an encouraging option for enhancing the efficacy of immunotherapeutics. These systems are designed to overcome immunosuppressive mechanisms in the TME to improve the efficacy of a therapy. This review will discuss different aspects of the TME and how they impede therapy success. Then, we will summarize recent developments in TME-modifying nanotherapeutics and the in vitro models utilized to facilitate these advances.
Collapse
Affiliation(s)
- Ryan Blanchard
- Department of Biomedical Engineering, Texas A&M University TX USA
| | - Isaac Adjei
- Department of Biomedical Engineering, Texas A&M University TX USA
| |
Collapse
|
18
|
Duan W, Xia S, Tang M, Lin M, Liu W, Wang Q. Targeting of endothelial cells in brain tumours. Clin Transl Med 2023; 13:e1433. [PMID: 37830128 PMCID: PMC10570772 DOI: 10.1002/ctm2.1433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/11/2023] [Accepted: 09/30/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Aggressive brain tumours, whether primary gliomas or secondary metastases, are characterised by hypervascularisation and are fatal. Recent research has emphasised the crucial involvement of endothelial cells (ECs) in all brain tumour genesis and development events, with various patterns and underlying mechanisms identified. MAIN BODY Here, we highlight recent advances in knowledge about the contributions of ECs to brain tumour development, providing a comprehensive summary including descriptions of interactions between ECs and tumour cells, the heterogeneity of ECs and new models for research on ECs in brain malignancies. We also discuss prospects for EC targeting in novel therapeutic approaches. CONCLUSION Interventions targeting ECs, as an adjunct to other therapies (e.g. immunotherapies, molecular-targeted therapies), have shown promising clinical efficacy due to the high degree of vascularisation in brain tumours. Developing precise strategies to target tumour-associated vessels based on the heterogeneity of ECs is expected to improve anti-vascular efficacy.
Collapse
Affiliation(s)
- Wenzhe Duan
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Shengkai Xia
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Mengyi Tang
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Manqing Lin
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Wenwen Liu
- Cancer Translational Medicine Research CenterThe Second HospitalDalian Medical UniversityDalianChina
| | - Qi Wang
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
- Cancer Translational Medicine Research CenterThe Second HospitalDalian Medical UniversityDalianChina
| |
Collapse
|
19
|
Dianat-Moghadam H, Nedaeinia R, Keshavarz M, Azizi M, Kazemi M, Salehi R. Immunotherapies targeting tumor vasculature: challenges and opportunities. Front Immunol 2023; 14:1226360. [PMID: 37727791 PMCID: PMC10506263 DOI: 10.3389/fimmu.2023.1226360] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/31/2023] [Indexed: 09/21/2023] Open
Abstract
Angiogenesis is a hallmark of cancer biology, and neoadjuvant therapies targeting either tumor vasculature or VEGF signaling have been developed to treat solid malignant tumors. However, these therapies induce complete vascular depletion leading to hypoxic niche, drug resistance, and tumor recurrence rate or leading to impaired delivery of chemo drugs and immune cell infiltration at the tumor site. Achieving a balance between oxygenation and tumor growth inhibition requires determining vascular normalization after treatment with a low dose of antiangiogenic agents. However, monotherapy within the approved antiangiogenic agents' benefits only some tumors and their efficacy improvement could be achieved using immunotherapy and emerging nanocarriers as a clinical tool to optimize subsequent therapeutic regimens and reduce the need for a high dosage of chemo agents. More importantly, combined immunotherapies and nano-based delivery systems can prolong the normalization window while providing the advantages to address the current treatment challenges within antiangiogenic agents. This review summarizes the approved therapies targeting tumor angiogenesis, highlights the challenges and limitations of current therapies, and discusses how vascular normalization, immunotherapies, and nanomedicine could introduce the theranostic potentials to improve tumor management in future clinical settings.
Collapse
Affiliation(s)
- Hassan Dianat-Moghadam
- Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mehdi Azizi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Kazemi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
20
|
Razi S, Haghparast A, Chodari Khameneh S, Ebrahimi Sadrabadi A, Aziziyan F, Bakhtiyari M, Nabi-Afjadi M, Tarhriz V, Jalili A, Zalpoor H. The role of tumor microenvironment on cancer stem cell fate in solid tumors. Cell Commun Signal 2023; 21:143. [PMID: 37328876 PMCID: PMC10273768 DOI: 10.1186/s12964-023-01129-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/15/2023] [Indexed: 06/18/2023] Open
Abstract
In the last few decades, the role of cancer stem cells in initiating tumors, metastasis, invasion, and resistance to therapies has been recognized as a potential target for tumor therapy. Understanding the mechanisms by which CSCs contribute to cancer progression can help to provide novel therapeutic approaches against solid tumors. In this line, the effects of mechanical forces on CSCs such as epithelial-mesenchymal transition, cellular plasticity, etc., the metabolism pathways of CSCs, players of the tumor microenvironment, and their influence on the regulating of CSCs can lead to cancer progression. This review focused on some of these mechanisms of CSCs, paving the way for a better understanding of their regulatory mechanisms and developing platforms for targeted therapies. While progress has been made in research, more studies will be required in the future to explore more aspects of how CSCs contribute to cancer progression. Video Abstract.
Collapse
Affiliation(s)
- Sara Razi
- Vira Pioneers of Modern Science (VIPOMS), Tehran, Iran
| | | | | | - Amin Ebrahimi Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACER, Tehran, Iran
- Cytotech and Bioinformatics Research Group, Tehran, Iran
| | - Fatemeh Aziziyan
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vahideh Tarhriz
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, P.O. Box 5163639888, Tabriz, Iran.
| | - Arsalan Jalili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACER, Tehran, Iran.
- Parvaz Research Ideas Supporter Institute, Tehran, Iran.
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
21
|
Cheng Y, Li S, Hou Y, Wang W, Wang K, Fu S, Yuan Y, Yang K, Ye X. Glioma-derived small extracellular vesicles induce pericyte-phenotype transition of glioma stem cells under hypoxic conditions. Cell Signal 2023:110754. [PMID: 37315748 DOI: 10.1016/j.cellsig.2023.110754] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Glioblastoma (GBM) is the most common and lethal primary brain tumor characterized by extensive vascularization. Anti-angiogenic therapy for this cancer offers the possibility of universal efficacy. However, preclinical and clinical studies suggest that anti-VEGF drug such as Bevacizumab actively promotes tumor invasion, which ultimately leads to a therapy-resistant and recurrent phenotype of GBMs. Whether Bevacizumab can improve survival over chemotherapy alone remains debated. Herein, we emphasized the importance of small extracellular vesicles (sEVs) internalization by glioma stem cells (GSCs) in giving rise to the failure of anti-angiogenic therapy in the treatment of GBMs and discovered a specific therapeutic target for this damaging disease. METHODS To experimentally prove that hypoxia condition promotes the release of GBM cells-derived sEVs, which could be taken up by the surrounding GSCs, we used an ultracentrifugation strategy to isolate GBM-derived sEVs under hypoxic or normoxic conditions, performed bioinformatics analysis and multidimensional molecular biology experiments, and established a xenograft mouse model. RESULTS The internalization of sEVs by GSCs was proved to promote tumor growth and angiogenesis through the pericyte-phenotype transition. Hypoxia-derived sEVs could efficiently deliver TGF-β1 to GSCs, thus resulting in the activation of the TGF-β signaling pathway and the consequent pericyte-phenotype transition. Specifically targeting GSC-derived pericyte using Ibrutinib can reverse the effects of GBM-derived sEVs and enhance the tumor-eradicating effects when combined with Bevacizumab. CONCLUSION This present study provides a new interpretation of the failure of anti-angiogenic therapy in the non-operative treatment of GBMs and discovers a promising therapeutic target for this intractable disease.
Collapse
Affiliation(s)
- Yue Cheng
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China
| | - Shijie Li
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China
| | - Yongying Hou
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China
| | - Weijun Wang
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China
| | - Ke Wang
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China
| | - Shihui Fu
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, Hainan Province, PR China
| | - Ye Yuan
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, PR China.
| | - Kaidi Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; Department of Oncology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, Hainan Province, PR China.
| | - Xiufeng Ye
- Institute of Pathology Department, Basic Medical College, Chongqing Medical University, Chongqing 400038, PR China.
| |
Collapse
|
22
|
Miyai M, Iwama T, Hara A, Tomita H. Exploring the Vital Link Between Glioma, Neuron, and Neural Activity in the Context of Invasion. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:669-679. [PMID: 37286277 DOI: 10.1016/j.ajpath.2023.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 06/09/2023]
Abstract
Because of their ability to infiltrate normal brain tissue, gliomas frequently evade microscopic surgical excision. The histologic infiltrative property of human glioma has been previously characterized as Scherer secondary structures, of which the perivascular satellitosis is a prospective target for anti-angiogenic treatment in high-grade gliomas. However, the mechanisms underlying perineuronal satellitosis remain unclear, and therapy remains lacking. Our knowledge of the mechanism underlying Scherer secondary structures has improved over time. New techniques, such as laser capture microdissection and optogenetic stimulation, have advanced our understanding of glioma invasion mechanisms. Although laser capture microdissection is a useful tool for studying gliomas that infiltrate the normal brain microenvironment, optogenetics and mouse xenograft glioma models have been extensively used in studies demonstrating the unique role of synaptogenesis in glioma proliferation and identification of potential therapeutic targets. Moreover, a rare glioma cell line is established that, when transplanted in the mouse brain, can replicate and recapitulate the human diffuse invasion phenotype. This review discusses the primary molecular causes of glioma, its histopathology-based invasive mechanisms, and the importance of neuronal activity and interactions between glioma cells and neurons in the brain microenvironment. It also explores current methods and models of gliomas.
Collapse
Affiliation(s)
- Masafumi Miyai
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan; Department of Neurosurgery, Hashima City Hospital, Gifu, Japan; Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan.
| |
Collapse
|
23
|
Advanced Bioinformatics Analysis and Genetic Technologies for Targeting Autophagy in Glioblastoma Multiforme. Cells 2023; 12:cells12060897. [PMID: 36980238 PMCID: PMC10047676 DOI: 10.3390/cells12060897] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
As the most malignant primary brain tumor in adults, a diagnosis of glioblastoma multiforme (GBM) continues to carry a poor prognosis. GBM is characterized by cytoprotective homeostatic processes such as the activation of autophagy, capability to confer therapeutic resistance, evasion of apoptosis, and survival strategy even in the hypoxic and nutrient-deprived tumor microenvironment. The current gold standard of therapy, which involves radiotherapy and concomitant and adjuvant chemotherapy with temozolomide (TMZ), has been a game-changer for patients with GBM, relatively improving both overall survival (OS) and progression-free survival (PFS); however, TMZ is now well-known to upregulate undesirable cytoprotective autophagy, limiting its therapeutic efficacy for induction of apoptosis in GBM cells. The identification of targets utilizing bioinformatics-driven approaches, advancement of modern molecular biology technologies such as clustered regularly interspaced short palindromic repeats (CRISPR)—CRISPR-associated protein (Cas9) or CRISPR-Cas9 genome editing, and usage of microRNA (miRNA)-mediated regulation of gene expression led to the selection of many novel targets for new therapeutic development and the creation of promising combination therapies. This review explores the current state of advanced bioinformatics analysis and genetic technologies and their utilization for synergistic combination with TMZ in the context of inhibition of autophagy for controlling the growth of GBM.
Collapse
|
24
|
Jeon H, Byun J, Kang H, Kim K, Lee E, Kim JH, Hong CK, Song SW, Kim YH, Chong S, Kim JH, Nam SJ, Park JE, Lee S. Proteomic analysis predicts anti-angiogenic resistance in recurred glioblastoma. J Transl Med 2023; 21:69. [PMID: 36732815 PMCID: PMC9893563 DOI: 10.1186/s12967-023-03936-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Recurrence is common in glioblastoma multiforme (GBM) because of the infiltrative, residual cells in the tumor margin. Standard therapy for GBM consists of surgical resection followed by chemotherapy and radiotherapy, but the median survival of GBM patients remains poor (~ 1.5 years). For recurrent GBM, anti-angiogenic treatment is one of the common treatment approaches. However, current anti-angiogenic treatment modalities are not satisfactory because of the resistance to anti-angiogenic agents in some patients. Therefore, we sought to identify novel prognostic biomarkers that can predict the therapeutic response to anti-angiogenic agents in patients with recurrent glioblastoma. METHODS We selected patients with recurrent GBM who were treated with anti-angiogenic agents and classified them into responders and non-responders to anti-angiogenic therapy. Then, we performed proteomic analysis using liquid-chromatography mass spectrometry (LC-MS) with formalin-fixed paraffin-embedded (FFPE) tissues obtained from surgical specimens. We conducted a gene-ontology (GO) analysis based on protein abundance in the responder and non-responder groups. Based on the LC-MS and GO analysis results, we identified potential predictive biomarkers for anti-angiogenic therapy and validated them in recurrent glioblastoma patients. RESULTS In the mass spectrometry-based approach, 4957 unique proteins were quantified with high confidence across clinical parameters. Unsupervised clustering analysis highlighted distinct proteomic patterns (n = 269 proteins) between responders and non-responders. The GO term enrichment analysis revealed a cluster of genes related to immune cell-related pathways (e.g., TMEM173, FADD, CD99) in the responder group, whereas the non-responder group had a high expression of genes related to nuclear replisome (POLD) and damaged DNA binding (ERCC2). Immunohistochemistry of these biomarkers showed that the expression levels of TMEM173 and FADD were significantly associated with the overall survival and progression-free survival of patients with recurrent GBM. CONCLUSIONS The candidate biomarkers identified in our protein analysis may be useful for predicting the clinical response to anti-angiogenic agents in patients with recurred GBM.
Collapse
Affiliation(s)
- Hanwool Jeon
- grid.413967.e0000 0001 0842 2126Translational Biomedical Research Group, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea ,grid.267370.70000 0004 0533 4667Department of Neurological Surgery, Brain Tumor Center, Asan Medical Center, University of Ulsan College of Medicine 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea ,grid.267370.70000 0004 0533 4667Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joonho Byun
- grid.267370.70000 0004 0533 4667Department of Neurological Surgery, Brain Tumor Center, Asan Medical Center, University of Ulsan College of Medicine 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea
| | - Hayeong Kang
- grid.267370.70000 0004 0533 4667Department of Neurological Surgery, Brain Tumor Center, Asan Medical Center, University of Ulsan College of Medicine 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea
| | - Kyunggon Kim
- grid.413967.e0000 0001 0842 2126Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Eunyeup Lee
- grid.413967.e0000 0001 0842 2126Translational Biomedical Research Group, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea ,grid.267370.70000 0004 0533 4667Department of Neurological Surgery, Brain Tumor Center, Asan Medical Center, University of Ulsan College of Medicine 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea ,grid.267370.70000 0004 0533 4667Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong Hoon Kim
- grid.267370.70000 0004 0533 4667Department of Neurological Surgery, Brain Tumor Center, Asan Medical Center, University of Ulsan College of Medicine 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea
| | - Chang Ki Hong
- grid.267370.70000 0004 0533 4667Department of Neurological Surgery, Brain Tumor Center, Asan Medical Center, University of Ulsan College of Medicine 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea
| | - Sang Woo Song
- grid.267370.70000 0004 0533 4667Department of Neurological Surgery, Brain Tumor Center, Asan Medical Center, University of Ulsan College of Medicine 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea
| | - Young-Hoon Kim
- grid.267370.70000 0004 0533 4667Department of Neurological Surgery, Brain Tumor Center, Asan Medical Center, University of Ulsan College of Medicine 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea
| | - Sangjoon Chong
- grid.267370.70000 0004 0533 4667Department of Neurological Surgery, Brain Tumor Center, Asan Medical Center, University of Ulsan College of Medicine 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea
| | - Jae Hyun Kim
- grid.267370.70000 0004 0533 4667Department of Neurological Surgery, Brain Tumor Center, Asan Medical Center, University of Ulsan College of Medicine 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea
| | - Soo Jeong Nam
- grid.267370.70000 0004 0533 4667Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Eun Park
- grid.267370.70000 0004 0533 4667Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seungjoo Lee
- Translational Biomedical Research Group, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea. .,Department of Neurological Surgery, Brain Tumor Center, Asan Medical Center, University of Ulsan College of Medicine 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea. .,Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
25
|
Frontiñán-Rubio J, Llanos-González E, García-Carpintero S, Peinado JR, Ballesteros-Yáñez I, Rayo MV, de la Fuente J, Pérez-García VM, Perez-Romasanta LA, Malumbres M, Alcaín FJ, Durán-Prado M. CoQ 10 reduces glioblastoma growth and infiltration through proteome remodeling and inhibition of angiogenesis and inflammation. Cell Oncol (Dordr) 2023; 46:65-77. [PMID: 36319818 PMCID: PMC9947058 DOI: 10.1007/s13402-022-00734-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
PURPOSE Most monotherapies available against glioblastoma multiforme (GBM) target individual hallmarks of this aggressive brain tumor with minimal success. In this article, we propose a therapeutic strategy using coenzyme Q10 (CoQ10) as a pleiotropic factor that crosses the blood-brain barrier and accumulates in cell membranes acting as an antioxidant, and in mitochondrial membranes as a regulator of cell bioenergetics and gene expression. METHODS Xenografts of U251 cells in nu/nu mice were used to assay tumor growth, hypoxia, angiogenesis, and inflammation. An orthotopic model was used to explore microglial infiltration, tumor growth, and invasion into the brain parenchyma. Cell proliferation, migration, invasion, proteome remodeling, and secretome were assayed in vitro. Conditioned media were used to assay angiogenesis, monocyte chemoattraction, and differentiation into macrophages in vitro. RESULTS CoQ10 treatment decreased tumor volume in xenografts and orthotopic models, although its effect on tumor cell proliferation was not direct. Tumors from mice treated with CoQ10 were less hypoxic and vascularized, having less infiltration from inflammatory cells. Treatment-induced downregulation of HIF-1α and NF-kB led to a complete remodeling of the tumor cells proteome and secretome, impacting angiogenesis, monocyte infiltration, and their differentiation into macrophages. Besides, tumor cell migration and invasion were drastically restricted by mechanisms involving modulation of the actin cytoskeleton and downregulation of matrix metalloproteases (MMPs). CONCLUSIONS CoQ10 has a pleiotropic effect on GBM growth, targeting several hallmarks simultaneously. Thus, its integration into current treatments of this fatal disease should be considered.
Collapse
Affiliation(s)
- Javier Frontiñán-Rubio
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Emilio Llanos-González
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Sonia García-Carpintero
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Juan Ramón Peinado
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Inmaculada Ballesteros-Yáñez
- EMAS Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Margarita Villar Rayo
- SaBio Research Group, Hunting Resources Research Institute (IREC), Ciudad Real, Spain
| | - José de la Fuente
- SaBio Research Group, Hunting Resources Research Institute (IREC), Ciudad Real, Spain
| | - Víctor M Pérez-García
- Laboratory of Mathematical Oncology, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Luis A Perez-Romasanta
- Radiology and Medicinal Physics, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Marcos Malumbres
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Francisco J Alcaín
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain.
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| | - Mario Durán-Prado
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071, Ciudad Real, Spain.
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Regional Centre for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| |
Collapse
|
26
|
Erbani J, Boon M, Akkari L. Therapy-induced shaping of the glioblastoma microenvironment: Macrophages at play. Semin Cancer Biol 2022; 86:41-56. [PMID: 35569742 DOI: 10.1016/j.semcancer.2022.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 01/27/2023]
Abstract
The intricate cross-talks between tumor cells and their microenvironment play a key role in cancer progression and resistance to treatment. In recent years, targeting pro-tumorigenic components of the tumor microenvironment (TME) has emerged as a tantalizing strategy to improve the efficacy of standard-of-care (SOC) treatments, particularly for hard-to-treat cancers such as glioblastoma. In this review, we explore how the distinct microenvironmental niches characteristic of the glioblastoma TME shape response to therapy. In particular, we delve into the interplay between tumor-associated macrophages (TAM) and glioblastoma cells within angiogenic and hypoxic niches, and interrogate their dynamic co-evolution upon SOC therapies that fuels malignancy. Resolving the complexity of therapy-induced alterations in the glioblastoma TME and their impact on disease relapse is a stepping stone to identify targetable pro-tumorigenic pathways and TAM subsets, and may open the way to efficient combination therapies that will improve clinical outcomes.
Collapse
Affiliation(s)
- Johanna Erbani
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Menno Boon
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Leila Akkari
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
27
|
Szklener K, Mazurek M, Wieteska M, Wacławska M, Bilski M, Mańdziuk S. New Directions in the Therapy of Glioblastoma. Cancers (Basel) 2022; 14:5377. [PMID: 36358795 PMCID: PMC9655599 DOI: 10.3390/cancers14215377] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma is the most common histologic type of all gliomas and contributes to 57.3% of all cases. Despite the standard management based on surgical resection and radiotherapy, it is related to poor outcome, with a 5-year relative survival rate below 6.9%. In order to improve the overall outcome for patients, the new therapeutic strategies are needed. Herein, we describe the current state of knowledge on novel targeted therapies in glioblastoma. Based on recent studies, we compared treatment efficacy measured by overall survival and progression-free survival in patients treated with selected potential antitumor drugs. The results of the application of the analyzed inhibitors are highly variable despite the encouraging conclusions of previous preclinical studies. This paper focused on drugs that target major glioblastoma kinases. As far, the results of some BRAF inhibitors are favorable. Vemurafenib demonstrated a long-term efficacy in clinical trials while the combination of dabrafenib and trametinib improves PFS compared with both vemurafenib and dabrafenib alone. There is no evidence that any MEK inhibitor is effective in monotherapy. According to the current state of knowledge, BRAF and MEK inhibition are more advantageous than BRAF inhibitor monotherapy. Moreover, mTOR inhibitors (especially paxalisib) may be considered a particularly important group. Everolimus demonstrated a partial response in a significant proportion of patients when combined with bevacizumab, however its actual role in the treatment is unclear. Neither nintedanib nor pemigatinib were efficient in treatment of GBM. Among the anti-VEGF drugs, bevacizumab monotherapy was a well-tolerated option, significantly associated with anti-GBM activity in patients with recurrent GBM. The efficacy of aflibercept and pazopanib in monotherapy has not been demonstrated. Apatinib has been proven to be effective and tolerable by a single clinical trial, but more research is needed. Lenvatinib is under trial. Finally, promising results from a study with regorafenib may be confirmed by the ongoing randomized AGILE trial. The studies conducted so far have provided a relatively wide range of drugs, which are at least well tolerated and demonstrated some efficacy in the randomized clinical trials. The comprehensive understanding of the molecular biology of gliomas promises to further improve the treatment outcomes of patients.
Collapse
Affiliation(s)
- Katarzyna Szklener
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 8 Jaczewski Street, 20-090 Lublin, Poland
| | - Marek Mazurek
- Department of Neurosurgery, Medical University of Lublin, 20-090 Lublin, Poland
| | - Małgorzata Wieteska
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 8 Jaczewski Street, 20-090 Lublin, Poland
| | - Monika Wacławska
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 8 Jaczewski Street, 20-090 Lublin, Poland
| | - Mateusz Bilski
- Department of Radiotherapy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Sławomir Mańdziuk
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 8 Jaczewski Street, 20-090 Lublin, Poland
| |
Collapse
|
28
|
Buccarelli M, Castellani G, Ricci-Vitiani L. Glioblastoma-Specific Strategies of Vascularization: Implications in Anti-Angiogenic Therapy Resistance. J Pers Med 2022; 12:jpm12101625. [PMID: 36294763 PMCID: PMC9604754 DOI: 10.3390/jpm12101625] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/09/2022] Open
Abstract
Angiogenesis has long been implicated as a crucial process in GBM growth and progression. GBM can adopt several strategies to build up its abundant and aberrant vasculature. Targeting GBM angiogenesis has gained more and more attention in anti-cancer therapy, and many strategies have been developed to interfere with this hallmark. However, recent findings reveal that the effects of anti-angiogenic treatments are temporally limited and that tumors become refractory to therapy and more aggressive. In this review, we summarize the GBM-associated neovascularization processes and their implication in drug resistance mechanisms underlying the transient efficacy of current anti-angiogenic therapies. Moreover, we describe potential strategies and perspectives to overcome the mechanisms adopted by GBM to develop resistance to anti-angiogenic therapy as new potential therapeutic approaches.
Collapse
Affiliation(s)
- Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
- Correspondence:
| |
Collapse
|
29
|
Lodi A, Pandey R, Chiou J, Bhattacharya A, Huang S, Pan X, Burgman B, Yi SS, Tiziani S, Brenner AJ. Circulating metabolites associated with tumor hypoxia and early response to treatment in bevacizumab-refractory glioblastoma after combined bevacizumab and evofosfamide. Front Oncol 2022; 12:900082. [PMID: 36226069 PMCID: PMC9549210 DOI: 10.3389/fonc.2022.900082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/07/2022] [Indexed: 12/02/2022] Open
Abstract
Glioblastomas (GBM) are the most common and aggressive form of primary malignant brain tumor in the adult population, and, despite modern therapies, patients often develop recurrent disease, and the disease remains incurable with median survival below 2 years. Resistance to bevacizumab is driven by hypoxia in the tumor and evofosfamide is a hypoxia-activated prodrug, which we tested in a phase 2, dual center (University of Texas Health Science Center in San Antonio and Dana Farber Cancer Institute) clinical trial after bevacizumab failure. Tumor hypoxic volume was quantified by 18F-misonidazole PET. To identify circulating metabolic biomarkers of tumor hypoxia in patients, we used a high-resolution liquid chromatography-mass spectrometry-based approach to profile blood metabolites and their specific enantiomeric forms using untargeted approaches. Moreover, to evaluate early response to treatment, we characterized changes in circulating metabolite levels during treatment with combined bevacizumab and evofosfamide in recurrent GBM after bevacizumab failure. Gamma aminobutyric acid, and glutamic acid as well as its enantiomeric form D-glutamic acid all inversely correlated with tumor hypoxia. Intermediates of the serine synthesis pathway, which is known to be modulated by hypoxia, also correlated with tumor hypoxia (phosphoserine and serine). Moreover, following treatment, lactic acid was modulated by treatment, likely in response to a hypoxia mediated modulation of oxidative vs glycolytic metabolism. In summary, although our results require further validation in larger patients’ cohorts, we have identified candidate metabolic biomarkers that could evaluate the extent of tumor hypoxia and predict the benefit of combined bevacizumab and evofosfamide treatment in GBM following bevacizumab failure.
Collapse
Affiliation(s)
- Alessia Lodi
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- *Correspondence: Alessia Lodi, ; Andrew J. Brenner,
| | - Renu Pandey
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Jennifer Chiou
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Ayon Bhattacharya
- Mays Cancer Center, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Shiliang Huang
- Mays Cancer Center, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Xingxin Pan
- Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, United States
| | - Brandon Burgman
- Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, United States
- Institute for Cellular and Molecular Biology (ICMB), College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
| | - S. Stephen Yi
- Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, United States
- Institute for Cellular and Molecular Biology (ICMB), College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, United States
- Oden Institute for Computational Engineering and Sciences (ICES), The University of Texas at Austin, Austin, TX, United States
| | - Stefano Tiziani
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, United States
- Institute for Cellular and Molecular Biology (ICMB), College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Andrew J. Brenner
- Mays Cancer Center, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- *Correspondence: Alessia Lodi, ; Andrew J. Brenner,
| |
Collapse
|
30
|
Takei J, Fukasawa N, Tanaka T, Yamamoto Y, Tamura R, Sasaki H, Akasaki Y, Kamata Y, Murahashi M, Shimoda M, Murayama Y. Impact of Neoadjuvant Bevacizumab on Neuroradiographic Response and Histological Findings Related to Tumor Stemness and the Hypoxic Tumor Microenvironment in Glioblastoma: Paired Comparison Between Newly Diagnosed and Recurrent Glioblastomas. Front Oncol 2022; 12:898614. [PMID: 35785200 PMCID: PMC9247463 DOI: 10.3389/fonc.2022.898614] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/17/2022] [Indexed: 12/03/2022] Open
Abstract
Background Previously, we reported that bevacizumab (Bev) produces histological and neuroradiographic alterations including changes in tumor oxygenation, induction of an immunosupportive tumor microenvironment, and inhibition of stemness. To confirm how those effects vary during Bev therapy, paired samples from the same patients with newly diagnosed glioblastoma (GBM) who received preoperative neoadjuvant Bev (neoBev) were investigated with immunohistochemistry before and after recurrence. Methods Eighteen samples from nine patients with newly diagnosed GBM who received preoperative neoBev followed by surgery and chemoradiotherapy and then autopsy or salvage surgery after recurrence were investigated. The expression of carbonic anhydrase 9 (CA9), hypoxia-inducible factor-1 alpha (HIF-1α), nestin, and Forkhead box M1 (FOXM1) was evaluated with immunohistochemistry. For comparison between neoBev and recurrent tumors, we divided the present cohort into two groups based on neuroradiographic response: good and poor responders (GR and PR, respectively) to Bev were defined by the tumor regression rate on T1-weighted images with gadolinium enhancement (T1Gd) and fluid-attenuated inversion recovery images. Patterns of recurrence after Bev therapy were classified as cT1 flare-up and T2-diffuse/T2-circumscribed. Furthermore, we explored the possibility of utilizing FOXM1 as a biomarker of survival in this cohort. Results A characteristic “pseudo-papillary”-like structure containing round-shaped tumor cells clustered adjacent to blood vessels surrounded by spindle-shaped tumor cells was seen only in recurrent tumors. Tumor cells at the outer part of the “pseudo-papillary” structure were CA9-positive (CA9+)/HIF-1α+, whereas cells at the inner part of this structure were CA9−/HIF-1α+ and nestin+/FOXM1+. CA9 and HIF-1α expression was lower in T1Gd-GR and decreased in the “T2-circumscribed/T2-diffuse” pattern compared with the “T1 flare-up” pattern, suggesting that tumor oxygenation was frequently observed in T1Gd-GR in initial tumors and in the “T2-circumscribed/T2-diffuse” pattern in recurrent tumors. FOXM1 low-expression tumors tended to have a better prognosis than that of FOXM1 high-expression tumors. Conclusion A “pseudo-papillary” structure was seen in recurrent GBM after anti-vascular endothelial growth factor therapy. Bev may contribute to tumor oxygenation, leading to inhibition of stemness and correlation with a neuroimaging response during Bev therapy. FOXM1 may play a role as a biomarker of survival during Bev therapy.
Collapse
Affiliation(s)
- Jun Takei
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| | - Nei Fukasawa
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Toshihide Tanaka
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
- Department of Neurosurgery, Jikei University School of Medicine Kashiwa Hospital, Kashiwa, Japan
- *Correspondence: Toshihide Tanaka,
| | - Yohei Yamamoto
- Department of Neurosurgery, Jikei University School of Medicine Daisan Hospital, Tokyo, Japan
| | - Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Hikaru Sasaki
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Yasuharu Akasaki
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| | - Yuko Kamata
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Mutsunori Murahashi
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Yuichi Murayama
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
31
|
Mosteiro A, Pedrosa L, Ferrés A, Diao D, Sierra À, González JJ. The Vascular Microenvironment in Glioblastoma: A Comprehensive Review. Biomedicines 2022; 10:biomedicines10061285. [PMID: 35740307 PMCID: PMC9219822 DOI: 10.3390/biomedicines10061285] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma multiforme, the deadliest primary brain tumor, is characterized by an excessive and aberrant neovascularization. The initial expectations raised by anti-angiogenic drugs were soon tempered due to their limited efficacy in improving the overall survival. Intrinsic resistance and escape mechanisms against anti-VEGF therapies evidenced that tumor angiogenesis is an intricate multifaceted phenomenon and that vessels not only support the tumor but exert indispensable interactions for resistance and spreading. This holistic review covers the essentials of the vascular microenvironment of glioblastoma, including the perivascular niche components, the vascular generation patterns and the implicated signaling pathways, the endothelial–tumor interrelation, and the interconnection between vessel aberrancies and immune disarrangement. The revised concepts provide novel insights into the preclinical models and the potential explanations for the failure of conventional anti-angiogenic therapies, leading to an era of new and combined anti-angiogenic-based approaches.
Collapse
Affiliation(s)
- Alejandra Mosteiro
- Department of Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (A.F.); (J.J.G.)
- Correspondence:
| | - Leire Pedrosa
- Laboratory of Experimental Oncological Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (L.P.); (D.D.); (À.S.)
| | - Abel Ferrés
- Department of Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (A.F.); (J.J.G.)
| | - Diouldé Diao
- Laboratory of Experimental Oncological Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (L.P.); (D.D.); (À.S.)
| | - Àngels Sierra
- Laboratory of Experimental Oncological Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (L.P.); (D.D.); (À.S.)
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - José Juan González
- Department of Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (A.F.); (J.J.G.)
- Laboratory of Experimental Oncological Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (L.P.); (D.D.); (À.S.)
| |
Collapse
|
32
|
MEOX2 Regulates the Growth and Survival of Glioblastoma Stem Cells by Modulating Genes of the Glycolytic Pathway and Response to Hypoxia. Cancers (Basel) 2022; 14:cancers14092304. [PMID: 35565433 PMCID: PMC9099809 DOI: 10.3390/cancers14092304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Glioblastoma is the most common incurable primary brain tumor in adults, typically leading to death within 15 months of diagnosis. Although there is an ongoing debate in the scientific community about the precise cellular origin of this tumor, glioblastoma stem cells (GSCs), which are able to self-renew, yield a full tumor mass, and determine chemo- and radio-resistance, are recognized to have a pivotal role. Our research aims to understand the role of the mesenchyme homeobox 2 (MEOX2) transcription factor in GSCs where it is strongly and specifically expressed. We have found that MEOX2 is indeed important for the survival of these cells. In fact, when we reduce its expression in two different GSC lines, they undergo a massive death accompanied by the inhibition of key genes of the glycolytic metabolism, the main source of energy for these cells. Our results reveal a novel function for MEOX2 in glioblastoma and suggest a mechanism through which GSCs may survive even in unfavorable conditions. Abstract The most widely accepted hypothesis for the development of glioblastoma suggests that glioblastoma stem-like cells (GSCs) are crucially involved in tumor initiation and recurrence as well as in the occurrence of chemo- and radio-resistance. Mesenchyme homeobox 2 (MEOX2) is a transcription factor overexpressed in glioblastoma, whose expression is negatively correlated with patient survival. Starting from our observation that MEOX2 expression is strongly enhanced in six GSC lines, we performed shRNA-mediated knock-down experiments in two different GSC lines and found that MEOX2 depletion resulted in the inhibition of cell growth and sphere-forming ability and an increase in apoptotic cell death. By a deep transcriptome analysis, we identified a core group of genes modulated in response to MEOX2 knock-down. Among these genes, the repressed ones are largely enriched in genes involved in the hypoxic response and glycolytic pathway, two strictly related pathways that contribute to the resistance of high-grade gliomas to therapies. An in silico study of the regulatory regions of genes differentially expressed by MEOX2 knock-down revealed that they mainly consisted of GC-rich regions enriched for Sp1 and Klf4 binding motifs, two main regulators of metabolism in glioblastoma. Our results show, for the first time, the involvement of MEOX2 in the regulation of genes of GSC metabolism, which is essential for the survival and growth of these cells.
Collapse
|
33
|
Tamai S, Ichinose T, Tsutsui T, Tanaka S, Garaeva F, Sabit H, Nakada M. Tumor Microenvironment in Glioma Invasion. Brain Sci 2022; 12:brainsci12040505. [PMID: 35448036 PMCID: PMC9031400 DOI: 10.3390/brainsci12040505] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/05/2023] Open
Abstract
A major malignant trait of gliomas is their remarkable infiltration capacity. When glioma develops, the tumor cells have already reached the distant part. Therefore, complete removal of the glioma is impossible. Recently, research on the involvement of the tumor microenvironment in glioma invasion has advanced. Local hypoxia triggers cell migration as an environmental factor. The transcription factor hypoxia-inducible factor (HIF) -1α, produced in tumor cells under hypoxia, promotes the transcription of various invasion related molecules. The extracellular matrix surrounding tumors is degraded by proteases secreted by tumor cells and simultaneously replaced by an extracellular matrix that promotes infiltration. Astrocytes and microglia become tumor-associated astrocytes and glioma-associated macrophages/microglia, respectively, in relation to tumor cells. These cells also promote glioma invasion. Interactions between glioma cells actively promote infiltration of each other. Surgery, chemotherapy, and radiation therapy transform the microenvironment, allowing glioma cells to invade. These findings indicate that the tumor microenvironment may be a target for glioma invasion. On the other hand, because the living body actively promotes tumor infiltration in response to the tumor, it is necessary to reconsider whether the invasion itself is friend or foe to the brain.
Collapse
|
34
|
Filimonova M, Shitova A, Soldatova O, Shevchenko L, Saburova A, Podosinnikova T, Surinova V, Shegay P, Kaprin A, Ivanov S, Filimonov A. Combination of NOS- and PDK-Inhibitory Activity: Possible Way to Enhance Antitumor Effects. Int J Mol Sci 2022; 23:ijms23020730. [PMID: 35054914 PMCID: PMC8775993 DOI: 10.3390/ijms23020730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/30/2021] [Accepted: 01/04/2022] [Indexed: 12/16/2022] Open
Abstract
We have previously demonstrated a high antitumor potential of NOS inhibitor T1023 (1-isobutanoyl-2-isopropylisothiourea hydrobromide): antitumor antiangiogenic activity in several animal tumor models and its ability to synergistically enhance the antitumor effects of bevacizumab, cyclophosphamide and γ-radiation. At the same time, rather rapid adaptation of experimental neoplasias to T1023 treatment was often observed. We attempted to enhance the antitumor activity of this NOS inhibitor by supplementing its molecular structure with a PDK-inhibiting fragment, dichloroacetate (DCA), which is capable of hypoxia-oriented toxic effects. We synthesized compound T1084 (1-isobutanoyl-2-isopropylisothiourea dichloroacetate). Its toxic properties, NOS-inhibiting and PDK-inhibiting activity in vivo, and antitumor activity on the mouse Ehrlich carcinoma model (SEC) were investigated in compare with T1023 and Na-DCA. We found that the change of the salt-forming acid from HBr to DCA does not increase the toxicity of 1-isobutanoyl-2-isopropylisothiourea salts, but significantly expands the biochemical and anti-tumor activity. New compound T1084 realizes in vivo NOS-inhibiting and PDK-inhibiting activity, quantitatively, at the level of the previous compounds, T1023 and Na-DCA. In two independent experiments on SEC model, a pronounced synergistic antitumor effect of T1084 was observed in compare with T1023 and Na-DCA at equimolar doses. There were no signs of SEC adaptation to T1084 treatment, while experimental neoplasia rapidly desensitized to the separate treatment of both T1023 and Na-DCA. The totality of the data obtained indicates that the combination of antiangiogenic and hypoxia-oriented toxic effects (in this case, within the molecular structure of the active substance) can increase the antitumor effect and suppress the development of hypoxic resistance of neoplasias. In general, the proposed approach can be used for the design of new anticancer agents.
Collapse
Affiliation(s)
- Marina Filimonova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.S.); (O.S.); (L.S.); (V.S.); (A.F.)
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (T.P.); (S.I.)
- Correspondence: ; Tel.: +7-903-8144013
| | - Anna Shitova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.S.); (O.S.); (L.S.); (V.S.); (A.F.)
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (T.P.); (S.I.)
| | - Olga Soldatova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.S.); (O.S.); (L.S.); (V.S.); (A.F.)
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (T.P.); (S.I.)
| | - Ljudmila Shevchenko
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.S.); (O.S.); (L.S.); (V.S.); (A.F.)
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (T.P.); (S.I.)
| | - Alina Saburova
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (T.P.); (S.I.)
| | - Tatjana Podosinnikova
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (T.P.); (S.I.)
| | - Valentina Surinova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.S.); (O.S.); (L.S.); (V.S.); (A.F.)
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (T.P.); (S.I.)
| | - Petr Shegay
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (P.S.); (A.K.)
| | - Andrey Kaprin
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (P.S.); (A.K.)
| | - Sergey Ivanov
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (T.P.); (S.I.)
| | - Alexander Filimonov
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.S.); (O.S.); (L.S.); (V.S.); (A.F.)
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (A.S.); (T.P.); (S.I.)
| |
Collapse
|
35
|
Du X, He Q, Zhang B, Li N, Zeng X, Li W. Diagnostic accuracy of diffusion-weighted imaging in differentiating glioma recurrence from posttreatment-related changes: a meta-analysis. Expert Rev Anticancer Ther 2021; 22:123-130. [PMID: 34727815 DOI: 10.1080/14737140.2022.2000396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Magnetic resonance imaging (MRI) is the most commonly used imaging method to evaluate glioma recurrence. However, conventional MRI has difficulty distinguishing glioma accurately. This study aimed to explore the value of diffusion weighted imaging (DWI) in evaluating glioma recurrence and post-treatment-related changes. RESEARCH DESIGN AND METHODS PubMed, Cochrane Library, Embase, Web of Science, China National Knowledge Infrastructure (CNKI), Wanfang Database and China Science and Technology Journal Database were extensively searched in accordance with inclusion criteria and exclusion criteria to obtain appropriate included studies. The quality of the included studies was evaluated using the Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) tool. Combined sensitivity and specificity and the area under the summary receiver operating characteristic curve (SROC) with the 95% confidence interval (CI) were calculated. RESULTS Seventeen high-quality studies were included. The combined sensitivity was 0.82 (95% CI: 0.76-0.87), the specificity was 0.83 (95% CI: 0.76-0.89), the positive likelihood ratio was 4.9 (95% CI: 3.2-7.5), the negative likelihood ratio was 0.21 (95% CI: 0.15-0.30), the diagnostic odds ratio was 23 (95%: CI 11-48), and the area under the SROC was 0.90 (95% CI: 0.87-0.92). CONCLUSIONS This meta-analysis suggests that DWI has high sensitivity, specificity and accuracy in differentiating glioma recurrence.
Collapse
Affiliation(s)
- Xiaoli Du
- Department of Radiology, Chengdu First People's Hospital, Chengdu, China
| | - Qian He
- Department of Radiology, Chengdu First People's Hospital, Chengdu, China
| | - Boli Zhang
- Department of Radiology, Chengdu First People's Hospital, Chengdu, China
| | - Na Li
- Department of Radiology, Chengdu First People's Hospital, Chengdu, China
| | - Xuewen Zeng
- Department of Radiology, Chengdu First People's Hospital, Chengdu, China
| | - Wenbo Li
- Department of Radiology, Chengdu First People's Hospital, Chengdu, China
| |
Collapse
|
36
|
Shi W, Qu C, Wang X, Liang X, Tan Y, Zhang H. Diffusion kurtosis imaging combined with dynamic susceptibility contrast-enhanced MRI in differentiating high-grade glioma recurrence from pseudoprogression. Eur J Radiol 2021; 144:109941. [PMID: 34735828 DOI: 10.1016/j.ejrad.2021.109941] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVES To compare the added value of diffusion kurtosis imaging (DKI) with the combination of dynamic susceptibility contrast-enhanced (DSC) MRI in differentiating glioma recurrence from pseudoprogression. METHODS Thirty-four patients with high-grade gliomas developing new and/or increasing enhanced lesions within six months after surgery and chemoradiotherapy were retrospectively analyzed. All patients were pathologically confirmed to have recurrent glioma (n = 22) or pseudoprogression (n = 12). The DKI and DSC MRI parameters were calculated based on the enhanced lesions on contrast-enhanced T1WI. ROC analysis was performed on significant variables to determine their diagnostic performance. Multivariate logistic regression was used to determine the best prediction model for discrimination. RESULTS The relative mean kurtosis (rMK), relative axial kurtosis (rKa), relative cerebral blood volume (rCBV), and relative mean transit time (rMTT) of glioma recurrence were higher than those of pseudoprogression (all, P < 0.05). The AUCs and diagnostic accuracy were 0.879 and 82.35% for rMK, 0.723 and 70.59% for rKa, 0.890 and 82.35% for rCBV, 0.765 and 73.53% for rMTT, respectively. A multivariate logistic regression model showed a significant contribution of rMK (P = 0.006) and rCBV (P = 0.009) as independent imaging classifiers for discrimination. The combined use of rMK and rCBV improved the AUC to 0.924 (P < 0.001) and the diagnostic accuracy to 88.24%. CONCLUSION DKI may be a valuable non-invasive tool in differentiating glioma recurrence from pseudoprogression, and its use in combination with DSC MRI can improve diagnostic performance in assessing treatment response compared with either technique alone.
Collapse
Affiliation(s)
- Wenwei Shi
- Department of Radiology, Zhongda Hospital, Southeast University, No. 87 Dingjiaqiao, Nanjing 210009, Jiangsu Province, PR China
| | - Chongxiao Qu
- Department of Pathology, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, No. 29 of Twin Towers Temple Street, Taiyuan 030012, Shanxi Province, PR China
| | - Xiaochun Wang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan 030001, Shanxi Province, PR China
| | - Xiao Liang
- Department of Radiology, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, No. 29 of Twin Towers Temple Street, Taiyuan 030012, Shanxi Province, PR China
| | - Yan Tan
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan 030001, Shanxi Province, PR China.
| | - Hui Zhang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan 030001, Shanxi Province, PR China.
| |
Collapse
|
37
|
Alvarez R, Mandal D, Chittiboina P. Canonical and Non-Canonical Roles of PFKFB3 in Brain Tumors. Cells 2021; 10:cells10112913. [PMID: 34831136 PMCID: PMC8616071 DOI: 10.3390/cells10112913] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 12/27/2022] Open
Abstract
PFKFB3 is a bifunctional enzyme that modulates and maintains the intracellular concentrations of fructose-2,6-bisphosphate (F2,6-P2), essentially controlling the rate of glycolysis. PFKFB3 is a known activator of glycolytic rewiring in neoplastic cells, including central nervous system (CNS) neoplastic cells. The pathologic regulation of PFKFB3 is invoked via various microenvironmental stimuli and oncogenic signals. Hypoxia is a primary inducer of PFKFB3 transcription via HIF-1alpha. In addition, translational modifications of PFKFB3 are driven by various intracellular signaling pathways that allow PFKFB3 to respond to varying stimuli. PFKFB3 synthesizes F2,6P2 through the phosphorylation of F6P with a donated PO4 group from ATP and has the highest kinase activity of all PFKFB isoenzymes. The intracellular concentration of F2,6P2 in cancers is maintained primarily by PFKFB3 allowing cancer cells to evade glycolytic suppression. PFKFB3 is a primary enzyme responsible for glycolytic tumor metabolic reprogramming. PFKFB3 protein levels are significantly higher in high-grade glioma than in non-pathologic brain tissue or lower grade gliomas, but without relative upregulation of transcript levels. High PFKFB3 expression is linked to poor survival in brain tumors. Solitary or concomitant PFKFB3 inhibition has additionally shown great potential in restoring chemosensitivity and radiosensitivity in treatment-resistant brain tumors. An improved understanding of canonical and non-canonical functions of PFKFB3 could allow for the development of effective combinatorial targeted therapies for brain tumors.
Collapse
Affiliation(s)
- Reinier Alvarez
- Department of Neurological Surgery, University of Colorado School of Medicine, Aurora, CO 80045, USA;
- Neurosurgery Unit for Pituitary and Inheritable Disorders, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA;
| | - Debjani Mandal
- Neurosurgery Unit for Pituitary and Inheritable Disorders, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA;
| | - Prashant Chittiboina
- Neurosurgery Unit for Pituitary and Inheritable Disorders, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA;
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA
- Correspondence:
| |
Collapse
|
38
|
Sulforaphane Causes Cell Cycle Arrest and Apoptosis in Human Glioblastoma U87MG and U373MG Cell Lines under Hypoxic Conditions. Int J Mol Sci 2021; 22:ijms222011201. [PMID: 34681862 PMCID: PMC8541491 DOI: 10.3390/ijms222011201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive primary brain tumor. The median survival rate from diagnosis ranges from 15 to 17 months because the tumor is resistant to most therapeutic strategies. GBM exhibits microvascular hyperplasia and pronounced necrosis triggered by hypoxia. Sulforaphane (SFN), an isothiocyanate derived from cruciferous vegetables, has already demonstrated the ability to inhibit cell proliferation, by provoking cell cycle arrest, and leading to apoptosis in many cell lines. In this study, we investigated the antineoplastic effects of SFN [20-80 μM for 48 h] in GBM cells under normoxic and hypoxic conditions. Cell viability assays, flow cytometry, and Western blot results revealed that SFN could induce apoptosis of GBM cells in a dose-dependent manner, under both conditions. In particular, SFN significantly induced caspase 3/7 activation and DNA fragmentation. Moreover, our results demonstrated that SFN suppressed GBM cells proliferation by arresting the cell cycle at the S-phase, also under hypoxic condition, and that these effects may be due in part to its ability to induce oxidative stress by reducing glutathione levels and to increase the phosphorylation of extracellular signal-regulated kinases (ERKs). Overall, we hypothesized that SFN treatment might serve as a potential therapeutic strategy, alone or in combination, against GBM.
Collapse
|
39
|
A vascularized tumoroid model for human glioblastoma angiogenesis. Sci Rep 2021; 11:19550. [PMID: 34599235 PMCID: PMC8486855 DOI: 10.1038/s41598-021-98911-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022] Open
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.
Collapse
|
40
|
Chen J, Lee H, Schmitt P, Choy CJ, Miller DM, Williams BJ, Bearer EL, Frieboes HB. Bioengineered Models to Study Microenvironmental Regulation of Glioblastoma Metabolism. J Neuropathol Exp Neurol 2021; 80:1012–1023. [PMID: 34524448 DOI: 10.1093/jnen/nlab092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Despite extensive research and aggressive therapies, glioblastoma (GBM) remains a central nervous system malignancy with poor prognosis. The varied histopathology of GBM suggests a landscape of differing microenvironments and clonal expansions, which may influence metabolism, driving tumor progression. Indeed, GBM metabolic plasticity in response to differing nutrient supply within these microenvironments has emerged as a key driver of aggressiveness. Additionally, emergent biophysical and biochemical interactions in the tumor microenvironment (TME) are offering new perspectives on GBM metabolism. Perivascular and hypoxic niches exert crucial roles in tumor maintenance and progression, facilitating metabolic relationships between stromal and tumor cells. Alterations in extracellular matrix and its biophysical characteristics, such as rigidity and topography, regulate GBM metabolism through mechanotransductive mechanisms. This review highlights insights gained from deployment of bioengineering models, including engineered cell culture and mathematical models, to study the microenvironmental regulation of GBM metabolism. Bioengineered approaches building upon histopathology measurements may uncover potential therapeutic strategies that target both TME-dependent mechanotransductive and biomolecular drivers of metabolism to tackle this challenging disease. Longer term, a concerted effort integrating in vitro and in silico models predictive of patient therapy response may offer a powerful advance toward tailoring of treatment to patient-specific GBM characteristics.
Collapse
Affiliation(s)
- Joseph Chen
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Hyunchul Lee
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Philipp Schmitt
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Caleb J Choy
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Donald M Miller
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Brian J Williams
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Elaine L Bearer
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| | - Hermann B Frieboes
- From the Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA (JC, CJC, HBF); Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA (JC, DMM, HBF); Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA (HL, BJW); Department of Medicine, University of Louisville, Louisville, Kentucky, USA (PS, DMM); Department of Radiation Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA (DMM, BJW, HBF); Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA (HBF); Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA (ELB)
| |
Collapse
|
41
|
Wang J, Ma X, Ma J. Identification of Four Enhancer-Associated Genes as Risk Signature for Diffuse Glioma Patients. J Mol Neurosci 2021; 72:410-419. [PMID: 34462884 DOI: 10.1007/s12031-021-01861-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 05/19/2021] [Indexed: 01/21/2023]
Abstract
The abnormal expressions of enhancer-associated genes have been reported to be correlated with poor prognosis of tumors, including glioblastoma (GBM). The objective of the current study is to predict prognosis by identifying enhancer-associated genes (EAGs). The profiles of genome-wide expressions of low-grade glioma (LGG) and GBM tissues in The Cancer Genome Atlas (TCGA) dataset were obtained to explore the expression patterns of EAGs in diffuse glioma. The capacity of prognosis prediction was validated by Rembrandt and GSE16011. Moreover, qPCR was utilized to confirm the effect of JQ1 and THZ1 on the EAGs. We detected 35 differentially expressed EAGs, which were predictive of overall survival. These candidate EAGs were then subjected to the multivariate cox regression analysis and were further scoped down to four signature genes, including TRAM2, SMAGP, KDELC2, and C7ORF25. A total of 662 patients were then stratified according to the expression levels of these four signature genes. The high-risk group accounted for poorer prognosis based on the Rembrandt and GSE16011 databases. The results of qPCR also demonstrated that the expression of the four EAGs could be abolished by JQ1 (bromodomain inhibitor) and THZ1 (CDK7 inhibitor) treatment. Our study not only highlights the potential role of EAGs, which can be used to improve clinical prognosis prediction in patients with diffuse glioma, but also sheds light on the specific biomarkers and therapeutic targets for diffuse glioma patients.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xin Ma
- Department of Gastroenterology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jie Ma
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
42
|
Moriconi C, Civita P, Neto C, Pilkington GJ, Gumbleton M. Caveolin-1, a Key Mediator Across Multiple Pathways in Glioblastoma and an Independent Negative Biomarker of Patient Survival. Front Oncol 2021; 11:701933. [PMID: 34490102 PMCID: PMC8417742 DOI: 10.3389/fonc.2021.701933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GB) remains an aggressive malignancy with an extremely poor prognosis. Discovering new candidate drug targets for GB remains an unmet medical need. Caveolin-1 (Cav-1) has been shown to act variously as both a tumour suppressor and tumour promoter in many cancers. The implications of Cav-1 expression in GB remains poorly understood. Using clinical and genomic databases we examined the relationship between tumour Cav-1 gene expression (including its spatial distribution) and clinical pathological parameters of the GB tumour and survival probability in a TCGA cohort (n=155) and CGGA cohort (n=220) of GB patients. High expression of Cav-1 represented a significant independent predictor of shortened survival (HR = 2.985, 5.1 vs 14.9 months) with a greater statistically significant impact in female patients and in the Proneural and Mesenchymal GB subtypes. High Cav-1 expression correlated with other factors associated with poor prognosis: IDH w/t status, high histological tumour grade and low KPS score. A total of 4879 differentially expressed genes (DEGs) in the GB tumour were found to correlate with Cav-1 expression (either positively or negatively). Pathway enrichment analysis highlighted an over-representation of these DEGs to certain biological pathways. Focusing on those that lie within a framework of epithelial to mesenchymal transition and tumour cell migration and invasion we identified 27 of these DEGs. We then examined the prognostic value of Cav-1 when used in combination with any of these 27 genes and identified a subset of combinations (with Cav-1) indicative of co-operative synergistic mechanisms of action. Overall, the work has confirmed Cav-1 can serve as an independent prognostic marker in GB, but also augment prognosis when used in combination with a panel of biomarkers or clinicopathologic parameters. Moreover, Cav-1 appears to be linked to many signalling entities within the GB tumour and as such this work begins to substantiate Cav-1 or its associated signalling partners as candidate target for GB new drug discovery.
Collapse
Affiliation(s)
- Chiara Moriconi
- School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
- Department of Pathology and Cell Biology, Columbia University, New York Presbyterian Hospital, New York, NY, United States
| | - Prospero Civita
- School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
- Brain Tumour Research Centre, School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Catia Neto
- School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Geoffrey J. Pilkington
- School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
- Brain Tumour Research Centre, School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
- Department of Basic and Clinical Neuroscience, Division of Neuroscience, Institute of Psychiatry & Neurology, King’s College London, London, United Kingdom
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
43
|
Mortezaee K. Normalization in tumor ecosystem: Opportunities and challenges. Cell Biol Int 2021; 45:2017-2030. [PMID: 34189798 DOI: 10.1002/cbin.11655] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/24/2021] [Accepted: 06/17/2021] [Indexed: 12/15/2022]
Abstract
Current research in cancer therapy aims to exploit efficient strategies to have long-lasting effects on tumors and to reduce or even revoke the chance of recurrence. Within the tumor stroma, O2 and nutrients are abnormally distributed between various cells (preferentially for supplying cancer cells), the immune contexture is abnormally positioned (permissive essentially for cells exhibiting tumor-promoting capacity), the fibroblast and fibrotic content is abnormally distributed (presence of both extracellular matrix [ECM] stiffening and ECM-degrading factors both for tumor-promoting purposes), and the tumor vasculature is abnormally orchestrated (for hindering drug delivery and increasing the chance of tumor metastasis). Resistance is actually an adaptive response to an imbalance in the tumor ecosystem; thus, the key consideration for effective cancer therapy is to bring back the normal status in this ecosystem so as to reach the desired durable outcome. Vascular normalization, metabolic modulation (glucose delivery in particular), balancing cellular dispersion, and balancing the pH rate and O2 delivery within the tumor microenvironment are suggested strategies to reverse abnormality within the tumor stroma.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
44
|
Talati P, El-Abtah M, Kim D, Dietrich J, Fu M, Wenke M, He J, Natheir SN, Vangel M, Rapalino O, Vaynrub A, Arrillaga-Romany I, Forst DA, Yen YF, Andronesi O, Kalpathy-Cramer J, Rosen B, Batchelor TT, Gonzalez RG, Gerstner ER, Ratai EM. MR spectroscopic imaging predicts early response to anti-angiogenic therapy in recurrent glioblastoma. Neurooncol Adv 2021; 3:vdab060. [PMID: 34131648 PMCID: PMC8193903 DOI: 10.1093/noajnl/vdab060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Background Determining failure to anti-angiogenic therapy in recurrent glioblastoma (GBM) (rGBM) remains a challenge. The purpose of the study was to assess treatment response to bevacizumab-based therapy in patients with rGBM using MR spectroscopy (MRS). Methods We performed longitudinal MRI/MRS in 33 patients with rGBM to investigate whether changes in N-acetylaspartate (NAA)/Choline (Cho) and Lactate (Lac)/NAA from baseline to subsequent time points after treatment can predict early failures to bevacizumab-based therapies. Results After stratifying based on 9-month survival, longer-term survivors had increased NAA/Cho and decreased Lac/NAA levels compared to shorter-term survivors. ROC analyses for intratumoral NAA/Cho correlated with survival at 1 day, 2 weeks, 8 weeks, and 16 weeks. Intratumoral Lac/NAA ROC analyses were predictive of survival at all time points tested. At the 8-week time point, 88% of patients with decreased NAA/Cho did not survive 9 months; furthermore, 90% of individuals with an increased Lac/NAA from baseline did not survive at 9 months. No other metabolic ratios tested significantly predicted survival. Conclusions Changes in metabolic levels of tumoral NAA/Cho and Lac/NAA can serve as early biomarkers for predicting treatment failure to anti-angiogenic therapy as soon as 1 day after bevacizumab-based therapy. The addition of MRS to conventional MR methods can provide better insight into how anti-angiogenic therapy affects tumor microenvironment and predict patient outcomes.
Collapse
Affiliation(s)
- Pratik Talati
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Mohamed El-Abtah
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Daniel Kim
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jorg Dietrich
- Harvard Medical School, Boston, Massachusetts, USA.,Massachusetts General Hospital, Cancer Center, Boston, Massachusetts, USA
| | - Melanie Fu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael Wenke
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Julian He
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Sharif N Natheir
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mark Vangel
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Otto Rapalino
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Anna Vaynrub
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Isabel Arrillaga-Romany
- Harvard Medical School, Boston, Massachusetts, USA.,Massachusetts General Hospital, Cancer Center, Boston, Massachusetts, USA
| | - Deborah A Forst
- Massachusetts General Hospital, Cancer Center, Boston, Massachusetts, USA
| | - Yi-Fen Yen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Ovidiu Andronesi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Jayashree Kalpathy-Cramer
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Bruce Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Tracy T Batchelor
- Harvard Medical School, Boston, Massachusetts, USA.,Massachusetts General Hospital, Cancer Center, Boston, Massachusetts, USA
| | - R Gilberto Gonzalez
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth R Gerstner
- Harvard Medical School, Boston, Massachusetts, USA.,Massachusetts General Hospital, Cancer Center, Boston, Massachusetts, USA
| | - Eva-Maria Ratai
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Rosińska S, Gavard J. Tumor Vessels Fuel the Fire in Glioblastoma. Int J Mol Sci 2021; 22:6514. [PMID: 34204510 PMCID: PMC8235363 DOI: 10.3390/ijms22126514] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
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.
Collapse
Affiliation(s)
- Sara Rosińska
- CRCINA, Inserm, CNRS, Université de Nantes, 44000 Nantes, France;
| | - Julie Gavard
- CRCINA, Inserm, CNRS, Université de Nantes, 44000 Nantes, France;
- Integrated Center for Oncology, ICO, 44800 St. Herblain, France
| |
Collapse
|
46
|
Khater AR, Abou-Antoun T. Mesenchymal Epithelial Transition Factor Signaling in Pediatric Nervous System Tumors: Implications for Malignancy and Cancer Stem Cell Enrichment. Front Cell Dev Biol 2021; 9:654103. [PMID: 34055785 PMCID: PMC8155369 DOI: 10.3389/fcell.2021.654103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022] Open
Abstract
Malignant nervous system cancers in children are the most devastating and worrisome diseases, specifically due to their aggressive nature and, in some cases, inoperable location in critical regions of the brain and spinal cord, and the impermeable blood-brain barrier that hinders delivery of pharmaco-therapeutic compounds into the tumor site. Moreover, the delicate developmental processes of the nervous system throughout the childhood years adds another limitation to the therapeutic modalities and doses used to treat these malignant cancers. Therefore, pediatric oncologists are charged with the daunting responsibility of attempting to deliver effective cures to these children, yet with limited doses of the currently available therapeutic options in order to mitigate the imminent neurotoxicity of radio- and chemotherapy on the developing nervous system. Various studies reported that c-Met/HGF signaling is affiliated with increased malignancy and stem cell enrichment in various cancers such as high-grade gliomas, high-risk medulloblastomas, and MYCN-amplified, high-risk neuroblastomas. Therapeutic interventions that are utilized to target c-Met signaling in these malignant nervous system cancers have shown benefits in basic translational studies and preclinical trials, but failed to yield significant clinical benefits in patients. While numerous pre-clinical data reported promising results with the use of combinatorial therapy that targets c-Met with other tumorigenic pathways, therapeutic resistance remains a problem, and long-term cures are rare. The possible mechanisms, including the overexpression and activation of compensatory tumorigenic mechanisms within the tumors or ineffective drug delivery methods that may contribute to therapeutic resistance observed in clinical trials are elaborated in this review.
Collapse
Affiliation(s)
- Amanda Rose Khater
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese American University, Byblos, Lebanon
| | - Tamara Abou-Antoun
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese American University, Byblos, Lebanon
| |
Collapse
|
47
|
Wirsching HG, Roth P, Weller M. A vasculature-centric approach to developing novel treatment options for glioblastoma. Expert Opin Ther Targets 2021; 25:87-100. [PMID: 33482697 DOI: 10.1080/14728222.2021.1881062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Glioblastoma is invariably deadly and is characterized by extensive vascularization and macrophage-dominant immunosuppression; nevertheless, anti-angiogenesis has so far failed to prolong overall survival of patients. Regardless of the problems in clinical development, the rationale for the application of anti-angiogenics in glioblastoma remains.Areas covered: Resistance to anti-angiogenics is discussed, including vessel co-option and amplification of hypoxic signaling in response to vessel destruction. The modulation of GSC and tumor-associated macrophages by dysfunctional tumor vessels and by hypoxia are outlined. Pharmacologic approaches to sensitizing glioblastomas to anti-angiogenics and evidence for the cooperation of anti-angiogenics with immunotherapies are summarized. Database search: https://pubmed.ncbi.nlm.nih.gov prior to December 12, 2020.Expert opinion: Despite drawbacks in the clinical development of vascular endothelial growth factor A (VEGF)-targeted agents, there is still rationale for the use of anti-angiogenics. The better understanding of vascular co-option and adverse effects of blood vessel destruction guides to improve strategies for vascular targeting. The pivotal role of the vasculature and of angiogenic factors such as VEGF for the induction and maintenance of immunosuppression in glioblastoma supports the use of anti-angiogenics in combination with immunotherapy. Proinflammatory repolarization of perivascular and perinecrotic tumor-associated macrophages is probably paramount for overcoming treatment resistance to virtually any treatment.
Collapse
Affiliation(s)
- Hans-Georg Wirsching
- Department of Neurology University Hospital and University of Zurich, Zurich, Switzerland
| | - Patrick Roth
- Department of Neurology University Hospital and University of Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
48
|
Qiao K, Liu Y, Xu Z, Zhang H, Zhang H, Zhang C, Chang Z, Lu X, Li Z, Luo C, Liu Y, Yang C, Sun T. RNA m6A methylation promotes the formation of vasculogenic mimicry in hepatocellular carcinoma via Hippo pathway. Angiogenesis 2021; 24:83-96. [PMID: 32920668 DOI: 10.1007/s10456-020-09744-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/01/2020] [Indexed: 12/17/2022]
Abstract
Vasculogenic mimicry (VM) formed by aggressive tumor cells to mimic vasculogenic networks plays an important role in the tumor malignancy of HCC. However, the pathogenesis underlying VM is complex and has not been fully defined. m6A is a common mRNA modification and has many biological effects. However, the relationship between m6A and VM remains unclear. In this research, we found that m6A methyltransferase METTL3 in HCC tissues was positively correlated with VM. The m6A level of mRNA significantly increased in 3D cultured cells treated with VEGFa and was related to VM formation. Transcriptome sequencing analysis of 3D cultured cells with knockdown Mettl3 showed that the Hippo pathway was involved in m6A-mediated VM formation. Further mechanism research indicated that the m6A modification of YAP1 mRNA affected the translation of YAP1 mRNA. In conclusion, m6A methylation plays a key role in VM formation in HCC. METTL3 and YAP1 could be potential therapeutic targets via impairing VM formation in anti-metastatic strategies.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Animals
- Carcinoma, Hepatocellular/blood supply
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Cell Line, Tumor
- Disease Progression
- Gene Expression Regulation, Neoplastic
- Gene Silencing
- Hippo Signaling Pathway
- Humans
- Liver Neoplasms/blood supply
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Methylation
- Methyltransferases/metabolism
- Mice, Inbred BALB C
- Mice, Nude
- Molecular Mimicry
- Prognosis
- Protein Biosynthesis
- Protein Serine-Threonine Kinases/metabolism
- RNA/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
- Transcription Factors/metabolism
- Xenograft Model Antitumor Assays
- YAP-Signaling Proteins
- Mice
Collapse
Affiliation(s)
- Kailiang Qiao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Yantao Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Zheng Xu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Haohao Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Heng Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Chao Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China
| | - Zhi Chang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Xinyan Lu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China
| | - Zhongwei Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China
| | - Ce Luo
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China
| | - Yanrong Liu
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining Medical University, No.89, Guhuai Road, Rencheng District, Jining, Shandong, China
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin, China.
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China.
| |
Collapse
|
49
|
Funakoshi Y, Hata N, Kuga D, Hatae R, Sangatsuda Y, Fujioka Y, Takigawa K, Mizoguchi M. Update on Chemotherapeutic Approaches and Management of Bevacizumab Usage for Glioblastoma. Pharmaceuticals (Basel) 2020; 13:E470. [PMID: 33339404 PMCID: PMC7766528 DOI: 10.3390/ph13120470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/15/2020] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma, the most common primary brain tumor in adults, has one of the most dismal prognoses in cancer. In 2009, bevacizumab was approved for recurrent glioblastoma in the USA. To evaluate the clinical impact of bevacizumab as a first-line drug for glioblastoma, two randomized clinical trials, AVAglio and RTOG 0825, were performed. Bevacizumab was found to improve progression-free survival (PFS) and was reported to be beneficial for maintaining patient performance status as an initial treatment. These outcomes led to bevacizumab approval in Japan in 2013 as an insurance-covered first-line drug for glioblastoma concurrently with its second-line application. However, prolongation of overall survival was not evinced in these clinical trials; hence, the clinical benefit of bevacizumab for newly diagnosed glioblastomas remains controversial. A recent meta-analysis of randomized controlled trials of bevacizumab combined with temozolomide in recurrent glioblastoma also showed an effect only on PFS, and the benefit of bevacizumab even for recurrent glioblastoma is controversial. Here, we discuss the clinical impact of bevacizumab for glioblastoma treatment by reviewing previous clinical trials and real-world evidence by focusing on Japanese experiences. Moreover, the efficacy and safety of bevacizumab are summarized, and we provide suggestions for updating the approaches and management of bevacizumab.
Collapse
Affiliation(s)
| | - Nobuhiro Hata
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan; (Y.F.); (D.K.); (R.H.); (Y.S.); (Y.F.); (K.T.); (M.M.)
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Despite significant improvement in understanding of molecular underpinnings driving glioblastoma, there is minimal improvement in overall survival of patients. This poor outcome is caused in part by traditional designs of early phase clinical trials, which focus on clinical assessments of drug toxicity and response. Window of opportunity trials overcome this shortcoming by assessing drug-induced on-target molecular alterations in post-treatment human tumor specimens. This article provides an overview of window of opportunity trials, including novel designs for incorporating biologic end points into early stage trials in context of brain tumors, and examples of successfully executed window of opportunity trials for glioblastoma.
Collapse
|