1
|
Aebisher D, Woźnicki P, Czarnecka-Czapczyńska M, Dynarowicz K, Szliszka E, Kawczyk-Krupka A, Bartusik-Aebisher D. Molecular Determinants for Photodynamic Therapy Resistance and Improved Photosensitizer Delivery in Glioma. Int J Mol Sci 2024; 25:8708. [PMID: 39201395 PMCID: PMC11354549 DOI: 10.3390/ijms25168708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Gliomas account for 24% of all the primary brain and Central Nervous System (CNS) tumors. These tumors are diverse in cellular origin, genetic profile, and morphology but collectively have one of the most dismal prognoses of all cancers. Work is constantly underway to discover a new effective form of glioma therapy. Photodynamic therapy (PDT) may be one of them. It involves the local or systemic application of a photosensitive compound-a photosensitizer (PS)-which accumulates in the affected tissues. Photosensitizer molecules absorb light of the appropriate wavelength, initiating the activation processes leading to the formation of reactive oxygen species and the selective destruction of inappropriate cells. Research focusing on the effective use of PDT in glioma therapy is already underway with promising results. In our work, we provide detailed insights into the molecular changes in glioma after photodynamic therapy. We describe a number of molecules that may contribute to the resistance of glioma cells to PDT, such as the adenosine triphosphate (ATP)-binding cassette efflux transporter G2, glutathione, ferrochelatase, heme oxygenase, and hypoxia-inducible factor 1. We identify molecular targets that can be used to improve the photosensitizer delivery to glioma cells, such as the epithelial growth factor receptor, neuropilin-1, low-density lipoprotein receptor, and neuropeptide Y receptors. We note that PDT can increase the expression of some molecules that reduce the effectiveness of therapy, such as Vascular endothelial growth factor (VEGF), glutamate, and nitric oxide. However, the scientific literature lacks clear data on the effects of PDT on many of the molecules described, and the available reports are often contradictory. In our work, we highlight the gaps in this knowledge and point to directions for further research that may enhance the efficacy of PDT in the treatment of glioma.
Collapse
Affiliation(s)
- David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of The Rzeszów University, 35-310 Rzeszów, Poland
| | - Paweł Woźnicki
- English Division Science Club, Medical College of The Rzeszów University, 35-310 Rzeszów, Poland;
| | - Magdalena Czarnecka-Czapczyńska
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Medical University of Silesia, Batorego 15 Street, 41-902 Bytom, Poland;
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of The University of Rzeszów, 35-310 Rzeszów, Poland;
| | - Ewelina Szliszka
- Department of Microbiology and Immunology, Medical University of Silesia, Poniatowskiego 15, 40-055 Katowice, Poland;
| | - Aleksandra Kawczyk-Krupka
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Medical University of Silesia, Batorego 15 Street, 41-902 Bytom, Poland;
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of The Rzeszów University, 35-310 Rzeszów, Poland;
| |
Collapse
|
2
|
Wada H, Shimauchi-Ohtaki H, Tosaka M, Kawashima T, Osawa T, Osawa S, Horiguchi K, Nakata S, Yoshimoto Y. Comparison of Early Postoperative Diffusion Weighted Magnetic Resonance Imaging Findings After Resection of Gliomas and Meningiomas. World Neurosurg 2024; 186:e296-e304. [PMID: 38548056 DOI: 10.1016/j.wneu.2024.03.126] [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: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
OBJECTIVE Glioma and meningioma require vastly different surgical approaches, even if only involving a simple craniotomy procedure. Diffusion weighted imaging (DWI) is useful for the postoperative evaluation of ischemic damage. The present study evaluated the expected but unproven differences in DWI findings. METHODS A total of 41 patients with meningiomas and 63 with gliomas met the inclusion criteria for adult cases with superficial lesions treated through simple supratentorial craniotomy. Postoperative DWI findings of DWI-positive rate, DWI-positive area type, and relationship with neurological deficits were evaluated. RESULTS The DWI-positive rate (P = 0.01) and the proportion of rim-type lesions (P < 0.01) were significantly more common in gliomas. Patients with meningiomas and DWI-positive areas presented with higher rates of new neurological deficits (P < 0.01), and patients with meningiomas on the left side were more likely to develop new neurological deficits (P = 0.02). Patients with gliomas tended to develop new deficits with larger DWI-positive area volumes (P = 0.04). CONCLUSIONS Postoperative early DWI-positive rate and rim-type lesions are more common after glioma resection than meningioma resection. Larger volumes of DWI-positive areas may be associated with postoperative neurological symptoms in gliomas. DWI-positive finding is less common after meningioma than glioma resection but more likely to be associated with new neurological symptoms. These differences are important for adequate postoperative DWI evaluation of common supratentorial brain tumors.
Collapse
Affiliation(s)
- Hajime Wada
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hiroya Shimauchi-Ohtaki
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.
| | - Masahiko Tosaka
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Takahiro Kawashima
- Department of Neurosurgery, Tatebayashi Kosei General Hospital, Tatebayashi, Gunma, Japan
| | - Tadashi Osawa
- Department of Neurosurgery, Hidaka Hospital, Takasaki, Gunma, Japan
| | - Sho Osawa
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo-Ku, Tokyo, Japan
| | - Keishi Horiguchi
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Satoshi Nakata
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yuhei Yoshimoto
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
3
|
Domka W, Bartusik-Aebisher D, Rudy I, Dynarowicz K, Pięta K, Aebisher D. Photodynamic therapy in brain cancer: mechanisms, clinical and preclinical studies and therapeutic challenges. Front Chem 2023; 11:1250621. [PMID: 38075490 PMCID: PMC10704472 DOI: 10.3389/fchem.2023.1250621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/14/2023] [Indexed: 09/13/2024] Open
Abstract
Cancer is a main cause of death and preferred methods of therapy depend on the type of tumor and its location. Gliomas are the most common primary intracranial tumor, accounting for 81% of malignant brain tumors. Although relatively rare, they cause significant mortality. Traditional methods include surgery, radiotherapy and chemotherapy; they also have significant associated side effects that cause difficulties related to tumor excision and recurrence. Photodynamic therapy has potentially fewer side effects, less toxicity, and is a more selective treatment, and is thus attracting increasing interest as an advanced therapeutic strategy. Photodynamic treatment of malignant glioma is considered to be a promising additional therapeutic option that is currently being extensively investigated in vitro and in vivo. This review describes the application of photodynamic therapy for treatment of brain cancer. The mechanism of photodynamic action is also described in this work as it applies to treatment of brain cancers such as glioblastoma multiforme. The pros and cons of photodynamic therapy for brain cancer are also discussed.
Collapse
Affiliation(s)
- Wojciech Domka
- Department of Otolaryngology, Medical College of the University of Rzeszów, Rzeszów, Poland
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of the University of Rzeszów, Rzeszów, Poland
| | - Izabela Rudy
- Students English Division Science Club, Medical College of the University of Rzeszów, Rzeszów, Poland
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of the University of Rzeszów, Rzeszów, Poland
| | - Karolina Pięta
- Students English Division Science Club, Medical College of the University of Rzeszów, Rzeszów, Poland
| | - David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of the University of Rzeszów, Rzeszów, Poland
| |
Collapse
|
4
|
Hsia T, Small JL, Yekula A, Batool SM, Escobedo AK, Ekanayake E, You DG, Lee H, Carter BS, Balaj L. Systematic Review of Photodynamic Therapy in Gliomas. Cancers (Basel) 2023; 15:3918. [PMID: 37568734 PMCID: PMC10417382 DOI: 10.3390/cancers15153918] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Over the last 20 years, gliomas have made up over 89% of malignant CNS tumor cases in the American population (NIH SEER). Within this, glioblastoma is the most common subtype, comprising 57% of all glioma cases. Being highly aggressive, this deadly disease is known for its high genetic and phenotypic heterogeneity, rendering a complicated disease course. The current standard of care consists of maximally safe tumor resection concurrent with chemoradiotherapy. However, despite advances in technology and therapeutic modalities, rates of disease recurrence are still high and survivability remains low. Given the delicate nature of the tumor location, remaining margins following resection often initiate disease recurrence. Photodynamic therapy (PDT) is a therapeutic modality that, following the administration of a non-toxic photosensitizer, induces tumor-specific anti-cancer effects after localized, wavelength-specific illumination. Its effect against malignant glioma has been studied extensively over the last 30 years, in pre-clinical and clinical trials. Here, we provide a comprehensive review of the three generations of photosensitizers alongside their mechanisms of action, limitations, and future directions.
Collapse
Affiliation(s)
- Tiffaney Hsia
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Julia L. Small
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
- Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Anudeep Yekula
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 554414, USA
| | - Syeda M. Batool
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ana K. Escobedo
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Emil Ekanayake
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Dong Gil You
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Bob S. Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
5
|
Miretti M, Graglia MAG, Suárez AI, Prucca CG. Photodynamic Therapy for glioblastoma: a light at the end of the tunnel. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2023. [DOI: 10.1016/j.jpap.2023.100161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
6
|
Ihata T, Nonoguchi N, Fujishiro T, Omura N, Kawabata S, Kajimoto Y, Wanibuchi M. The effect of hypoxia on photodynamic therapy with 5-aminolevulinic acid in malignant gliomas. Photodiagnosis Photodyn Ther 2022; 40:103056. [PMID: 35944845 DOI: 10.1016/j.pdpdt.2022.103056] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/24/2022] [Accepted: 08/05/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Glioblastoma (GBM) is a high-grade, poor prognosis tumor that is resistant to standard treatment. The presence of a small number of glioma stem cells (GSCs) surviving in the harsh microenvironment is responsible for their refractoriness. This study aimed to investigate the effect of a hypoxic environment on the sensitivity of GSCs to photodynamic therapy with 5-aminolevulinic acid (ALA-PDT). MATERIALS AND METHODS Six human GSC lines, Mesenchymal types HGG13, HGG30, HGG1123, and Proneural types HGG146, HGG157, HGG528, were divided into two groups: normoxia (O2 21%)-cultured cells (Normoxia-GSCs), and hypoxia (O2 5%)-cultured cells (Hypoxia-GSCs). To compare the effects of different oxygen partial pressures on photoporphyrin Ⅸ (PpⅨ) biosynthetic activity, PpⅨ biosynthetic enzyme and transporter expression levels were examined by qRT-PCR; the intracellular PpⅨ concentration was determined using flow cytometry. Additionally, the sensitivity of these two groups of cells to ALA-PDT was evaluated in vitro. RESULTS Hypoxia-GSCs showed higher mRNA levels of FECH (ferrochelatase), which is required for iron synthesis to convert PpⅨ to heme, compared with Normoxia-GSCs. Flow cytometry revealed that the accumulation of PpⅨ in Hypoxia-GSCs reduced upon incubation with ALA. However, Hypoxia-GSCs showed less reduction in sensitivity to ALA-PDT than Normoxia-GSCs. CONCLUSION Hypoxia-GSCs had lower intracellular PpⅨ accumulation than Normoxia-GSCs due to increased gene expression of FECH, and that their sensitivity to ALA-PDT was reduced less, despite accumulating lower concentrations of PpⅨ. ALA-PDT is a potentially effective therapy for hypoxia-tolerant GSCs that exist in hypoxia at 5% oxygen concentration.
Collapse
Affiliation(s)
- Tomohiro Ihata
- Department of Neurosurgery and Endovascular Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686, Japan
| | - Naosuke Nonoguchi
- Department of Neurosurgery and Endovascular Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686, Japan.
| | - Takahiro Fujishiro
- Department of Neurosurgery, Tanabe Neurosurgical Hospital, Fujiidera, Osaka, Japan
| | - Naoki Omura
- Department of Neurosurgery and Endovascular Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686, Japan
| | - Shinji Kawabata
- Department of Neurosurgery and Endovascular Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686, Japan
| | - Yoshinaga Kajimoto
- Department of Neurosurgery and Endovascular Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686, Japan
| | - Masahiko Wanibuchi
- Department of Neurosurgery and Endovascular Neurosurgery, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-Machi, Takatsuki, Osaka 569-8686, Japan
| |
Collapse
|
7
|
Alva R, Gardner GL, Liang P, Stuart JA. Supraphysiological Oxygen Levels in Mammalian Cell Culture: Current State and Future Perspectives. Cells 2022; 11:3123. [PMID: 36231085 PMCID: PMC9563760 DOI: 10.3390/cells11193123] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Most conventional incubators used in cell culture do not regulate O2 levels, making the headspace O2 concentration ~18%. In contrast, most human tissues are exposed to 2-6% O2 (physioxia) in vivo. Accumulating evidence has shown that such hyperoxic conditions in standard cell culture practices affect a variety of biological processes. In this review, we discuss how supraphysiological O2 levels affect reactive oxygen species (ROS) metabolism and redox homeostasis, gene expression, replicative lifespan, cellular respiration, and mitochondrial dynamics. Furthermore, we present evidence demonstrating how hyperoxic cell culture conditions fail to recapitulate the physiological and pathological behavior of tissues in vivo, including cases of how O2 alters the cellular response to drugs, hormones, and toxicants. We conclude that maintaining physioxia in cell culture is imperative in order to better replicate in vivo-like tissue physiology and pathology, and to avoid artifacts in research involving cell culture.
Collapse
Affiliation(s)
- Ricardo Alva
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | | | | | | |
Collapse
|
8
|
Aiyappa-Maudsley R, Chalmers AJ, Parsons JL. Factors affecting the radiation response in glioblastoma. Neurooncol Adv 2022; 4:vdac156. [PMID: 36325371 PMCID: PMC9617255 DOI: 10.1093/noajnl/vdac156] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Glioblastoma (GBM) is a highly invasive primary brain tumor in adults with a 5-year survival rate of less than 10%. Conventional radiotherapy with photons, along with concurrent and adjuvant temozolomide, is the mainstay for treatment of GBM although no significant improvement in survival rates has been observed over the last 20 years. Inherent factors such as tumor hypoxia, radioresistant GBM stem cells, and upregulated DNA damage response mechanisms are well established as contributing to treatment resistance and tumor recurrence. While it is understandable that efforts have focused on targeting these factors to overcome this phenotype, there have also been striking advances in precision radiotherapy techniques, including proton beam therapy and carbon ion radiotherapy (CIRT). These enable higher doses of radiation to be delivered precisely to the tumor, while minimizing doses to surrounding normal tissues and organs at risk. These alternative radiotherapy techniques also benefit from increased biological effectiveness, particularly in the case of CIRT. Although not researched extensively to date, combining these new radiation modalities with radio-enhancing agents may be particularly effective in improving outcomes for patients with GBM.
Collapse
Affiliation(s)
- Radhika Aiyappa-Maudsley
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK
| | - Anthony J Chalmers
- Institute of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jason L Parsons
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK
- Clatterbridge Cancer Centre NHS Foundation Trust, Clatterbridge Road, Bebington, CH63 4JY, UK
| |
Collapse
|
9
|
Fontana LC, Pinto JG, Vitorio GDS, Ferreira I, Pacheco-Soares C, Mamone LA, Strixino JF. Photodynamic effect of protoporphyrin IX in gliosarcoma 9l/lacZ cell line. Photodiagnosis Photodyn Ther 2021; 37:102669. [PMID: 34863947 DOI: 10.1016/j.pdpdt.2021.102669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 11/29/2022]
Abstract
Photodynamic Therapy (PDT) is an oncologic treatment, producing reactive oxygen species (ROS) to induce the death of cancer cells. This study aimed to evaluate the action of PDT on gliosarcoma cells, using protoporphyrin IX as PS by incubation with the precursor aminolevulinic acid (ALA). An LED device was used with a light dose of 10 J/cm². The success of the therapy proved to be dependent on the concentration of ALA, and an incubation time of 4 h required for an effective response. Cell death was prevalent due to necrosis when assessed 18 h post-PDT. ALA proved to be an option to PDT in cells of the 9 L/lacZ, with the protocol tested.
Collapse
Affiliation(s)
- Letícia Corrêa Fontana
- Photobiology Applied to Health - Universidade do Vale do Paraíba. Av. Shishima Hifumi, 2911, Urbanova, São José dos Campos, São Paulo
| | - Juliana Guerra Pinto
- Photobiology Applied to Health - Universidade do Vale do Paraíba. Av. Shishima Hifumi, 2911, Urbanova, São José dos Campos, São Paulo
| | - Gabrielle Dos Santos Vitorio
- Photobiology Applied to Health - Universidade do Vale do Paraíba. Av. Shishima Hifumi, 2911, Urbanova, São José dos Campos, São Paulo
| | - Isabelle Ferreira
- Photobiology Applied to Health - Universidade do Vale do Paraíba. Av. Shishima Hifumi, 2911, Urbanova, São José dos Campos, São Paulo
| | - Cristina Pacheco-Soares
- Laboratory of Cellular Dynamics - Universidade do Vale do Paraíba. Av. Shishima Hifumi, 2911, Urbanova, São José dos Campos, São Paulo
| | - Leandro Ariel Mamone
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), CONICET and Hospital de Clínicas José de San Martín, Universidad de Buenos Aires. Córdoba 2351 1er subsuelo, Ciudad de Buenos Aires CP1120AAF, Argentina
| | - Juliana Ferreira Strixino
- Photobiology Applied to Health - Universidade do Vale do Paraíba. Av. Shishima Hifumi, 2911, Urbanova, São José dos Campos, São Paulo.
| |
Collapse
|
10
|
Takahashi J, Nagasawa S, Doi M, Takahashi M, Narita Y, Yamamoto J, Ikemoto MJ, Iwahashi H. In Vivo Study of the Efficacy and Safety of 5-Aminolevulinic Radiodynamic Therapy for Glioblastoma Fractionated Radiotherapy. Int J Mol Sci 2021; 22:ijms22189762. [PMID: 34575921 PMCID: PMC8470662 DOI: 10.3390/ijms22189762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/20/2022] Open
Abstract
To treat malignant glioma, standard fractionated radiotherapy (RT; 60 Gy/30 fractions over 6 weeks) was performed post-surgery in combination with temozolomide to improve overall survival. Malignant glioblastoma recurrence rate is extremely high, and most recurrent tumors originate from the excision cavity in the high-dose irradiation region. In our previous study, protoporphyrin IX physicochemically enhanced reactive oxygen species generation by ionizing radiation and combined treatment with 5-aminolevulinic acid (5-ALA) and ionizing radiation, while radiodynamic therapy (RDT) improved tumor growth suppression in vivo in a melanoma mouse model. We examined the effect of 5-ALA RDT on the standard fractionated RT protocol using U251MG- or U87MG-bearing mice. 5-ALA was orally administered at 60 or 120 mg/kg, 4 h prior to irradiation. In both models, combined treatment with 5-ALA slowed tumor progression and promoted regression compared to treatment with ionizing radiation alone. The standard fractionated RT protocol of 60 Gy in 30 fractions with oral administration of 120 and 240 mg/kg 5-ALA, the human equivalent dose of photodynamic diagnosis, revealed no significant increase in toxicity to normal skin or brain tissue compared to ionizing radiation alone. Thus, RDT is expected to enhance RT treatment of glioblastoma without severe toxicity under clinically feasible conditions.
Collapse
Affiliation(s)
- Junko Takahashi
- Graduate School of Information, Production and Systems, Waseda University, Fukuoka 808-0135, Japan
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki 305-8566, Japan;
- Correspondence: ; Tel.: +81-936-92-5154
| | - Shinsuke Nagasawa
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan;
| | - Motomichi Doi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki 305-8566, Japan;
| | - Masamichi Takahashi
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan; (M.T.); (Y.N.)
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan; (M.T.); (Y.N.)
| | - Junkoh Yamamoto
- Department of Neurosurgery, University of Occupational and Environmental Health, Fukuoka 807-8555, Japan;
| | - Mitsushi J. Ikemoto
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki 305-8566, Japan;
| | - Hitoshi Iwahashi
- The United Graduate School of Agricultural Science, Gifu University, Gifu 501-1193, Japan;
| |
Collapse
|
11
|
Systematic Review and Meta-Analysis of In Vitro Anti-Human Cancer Experiments Investigating the Use of 5-Aminolevulinic Acid (5-ALA) for Photodynamic Therapy. Pharmaceuticals (Basel) 2021; 14:ph14030229. [PMID: 33800109 PMCID: PMC8000125 DOI: 10.3390/ph14030229] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 12/11/2022] Open
Abstract
5-Aminolevulinic acid (5-ALA) is an amino acid derivative and a precursor of protoporphyrin IX (PpIX). The photophysical feature of PpIX is clinically used in photodynamic diagnosis (PDD) and photodynamic therapy (PDT). These clinical applications are potentially based on in vitro cell culture experiments. Thus, conducting a systematic review and meta-analysis of in vitro 5-ALA PDT experiments is meaningful and may provide opportunities to consider future perspectives in this field. We conducted a systematic literature search in PubMed to summarize the in vitro 5-ALA PDT experiments and calculated the effectiveness of 5-ALA PDT for several cancer cell types. In total, 412 articles were identified, and 77 were extracted based on our inclusion criteria. The calculated effectiveness of 5-ALA PDT was statistically analyzed, which revealed a tendency of cancer-classification-dependent sensitivity to 5-ALA PDT, and stomach cancer was significantly more sensitive to 5-ALA PDT compared with cancers of different origins. Based on our analysis, we suggest a standardized in vitro experimental protocol for 5-ALA PDT.
Collapse
|
12
|
Traylor JI, Pernik MN, Sternisha AC, McBrayer SK, Abdullah KG. Molecular and Metabolic Mechanisms Underlying Selective 5-Aminolevulinic Acid-Induced Fluorescence in Gliomas. Cancers (Basel) 2021; 13:cancers13030580. [PMID: 33540759 PMCID: PMC7867275 DOI: 10.3390/cancers13030580] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary 5-aminolevulinic acid (5-ALA) is a medication that produces fluorescence in certain cancers, which enables surgeons to visualize tumor margins during surgery. Gliomas are brain tumors that can be difficult to fully resect due to their infiltrative nature. In this review we explored what is known about the mechanism of 5-ALA, recent discoveries that increase our understanding of that mechanism, and potential targets to increase fluorescence in lower grade gliomas. Abstract 5-aminolevulinic acid (5-ALA) is a porphyrin precursor in the heme synthesis pathway. When supplied exogenously, certain cancers consume 5-ALA and convert it to the fluorogenic metabolite protoporphyrin IX (PpIX), causing tumor-specific tissue fluorescence. Preoperative administration of 5-ALA is used to aid neurosurgical resection of high-grade gliomas such as glioblastoma, allowing for increased extent of resection and progression free survival for these patients. A subset of gliomas, especially low-grade tumors, do not accumulate PpIX intracellularly or readily fluoresce upon 5-ALA administration, making gross total resection difficult to achieve in diffuse lesions. We review existing literature on 5-ALA metabolism and PpIX accumulation to explore potential mechanisms of 5-ALA-induced glioma tissue fluorescence. Targeting the heme synthesis pathway and understanding its dysregulation in malignant tissues could aid the development of adjunct therapies to increase intraoperative fluorescence after 5-ALA treatment.
Collapse
Affiliation(s)
- Jeffrey I. Traylor
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (J.I.T.); (M.N.P.)
| | - Mark N. Pernik
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (J.I.T.); (M.N.P.)
| | - Alex C. Sternisha
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Samuel K. McBrayer
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Correspondence: (S.K.M.); (K.G.A.); Tel.: +1-(214)-648-3730 (S.K.M.); +1-(214)-645-2300 (K.G.A.)
| | - Kalil G. Abdullah
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (J.I.T.); (M.N.P.)
- Correspondence: (S.K.M.); (K.G.A.); Tel.: +1-(214)-648-3730 (S.K.M.); +1-(214)-645-2300 (K.G.A.)
| |
Collapse
|
13
|
Chelakkot VS, Liu K, Yoshioka E, Saha S, Xu D, Licursi M, Dorward A, Hirasawa K. MEK reduces cancer-specific PpIX accumulation through the RSK-ABCB1 and HIF-1α-FECH axes. Sci Rep 2020; 10:22124. [PMID: 33335181 PMCID: PMC7747616 DOI: 10.1038/s41598-020-79144-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023] Open
Abstract
The efficacy of aminolevulinic acid (5-ALA)-based photodynamic diagnosis (5-ALA-PDD) and photodynamic therapy (5-ALA-PDT) is dependent on 5-ALA-induced cancer-specific accumulation of protoporphyrin IX (PpIX). We previously reported that inhibition of oncogenic Ras/MEK increases PpIX accumulation in cancer cells by reducing PpIX efflux through ATP-binding cassette sub-family B member 1 (ABCB1) and ferrochelatase (FECH)-catalysed PpIX conversion to haem. Here, we sought to identify the downstream pathways of Ras/MEK involved in the regulation of PpIX accumulation via ABCB1 and FECH. First, we demonstrated that Ras/MEK activation reduced PpIX accumulation in RasV12-transformed NIH3T3 cells and HRAS transgenic mice. Knockdown of p90 ribosomal S6 kinases (RSK) 2, 3, or 4 increased PpIX accumulation in RasV12-transformed NIH3T3 cells. Further, treatment with an RSK inhibitor reduced ABCB1 expression and increased PpIX accumulation. Moreover, HIF-1α expression was reduced when RasV12-transformed NIH3T3 cells were treated with a MEK inhibitor, demonstrating that HIF-1α is a downstream element of MEK. HIF-1α inhibition decreased FECH activity and increased PpIX accumulation. Finally, we demonstrated the involvement of RSKs and HIF-1α in the regulation of PpIX accumulation in human cancer cell lines. These results demonstrate that the RSK-ABCB1 and HIF-1α-FECH axes are the downstream pathways of Ras/MEK involved in the regulation of PpIX accumulation.
Collapse
Affiliation(s)
- Vipin Shankar Chelakkot
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Kaiwen Liu
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Ema Yoshioka
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Shaykat Saha
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Danyang Xu
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Maria Licursi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Ann Dorward
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Kensuke Hirasawa
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada.
| |
Collapse
|
14
|
Zhu H, Wang H, Huang Q, Liu Q, Guo Y, Lu J, Li X, Xue C, Han Q. Transcriptional Repression of p53 by PAX3 Contributes to Gliomagenesis and Differentiation of Glioma Stem Cells. Front Mol Neurosci 2018; 11:187. [PMID: 29937714 PMCID: PMC6003214 DOI: 10.3389/fnmol.2018.00187] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 05/14/2018] [Indexed: 12/31/2022] Open
Abstract
Although there are available therapies as surgery, chemotherapy and radiation, glioblastoma (GBM) still has been considered as the most common and overwhelming primary tumor of brain. In GBM, the brain glioma stem cells (BGSCs) were identified and played a crucial role in resistance of GBM to conventional therapies described above. PAX3 was previously identified by our group as a diagnostic/prognostic marker and a therapeutic regulator in the therapy of GBM. Here, we hypothesized PAX3/p53 axis promoted the process of differentiation, regulating to the cancer stem cell properties, such as proliferation and migration. The correlation between PAX3 and p53 in GBM were first clarified. Immunofluorescence of p53 was shown activated following BGSCs differentiation. We further identified that PAX3 might specifically bind to the promoter of p53 gene, and transcriptionally repressed p53 expression. ChIP assay further confirmed that PAX3/p53 axis regulated the differentiation process of BGSCs. Then, the function of PAX3 in BGSCs were sequentially investigated in vitro and in vivo. Ectopic PAX3 expression promoted BGSCs growth and migration while PAX3 knockdown suppressed BGSCs growth, migration in vitro and in vivo. Similar to PAX3 overexpression, p53 inhibition also showed increase in growth and migration of differentiated BGSCs. Regarding the functional interaction between PAX3 and p53, PAX3 knockdown-mediated decrease in proliferation was partially rescued by p53 inhibition. Hypoxia significantly promoted the migration potential of BGSCs. In addition, hypoxia inducible factor-1α (HIF-1α) might be a potential upstream regulator of PAX3 in differentiated BGSCs under hypoxia. Our work may provide a supplementary mechanism in regulation of the BGSCs differentiation and their functions, which should provide novel therapeutic targets for GBM in future.
Collapse
Affiliation(s)
- Hui Zhu
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Hongkui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Qingfeng Huang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Qianqian Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yibing Guo
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Jingjing Lu
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaohong Li
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Chengbin Xue
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Qianqian Han
- National Institute for Food and Drug Control, Beijing, China
| |
Collapse
|
15
|
HIF1α regulates single differentiated glioma cell dedifferentiation to stem-like cell phenotypes with high tumorigenic potential under hypoxia. Oncotarget 2018; 8:28074-28092. [PMID: 28427209 PMCID: PMC5438632 DOI: 10.18632/oncotarget.15888] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 02/20/2017] [Indexed: 01/24/2023] Open
Abstract
The standard treatment for Glioblastoma multiforme (GBM) is surgical resection and subsequent radiotherapy and chemotherapy. Surgical resection of GBM is typically restricted because of its invasive growth, which results in residual tumor cells including glioma stem cells (GSCs) and differentiated cells. Recurrence has been previously thought to occur as a result of these GSCs, and hypoxic microenvironment maintains the GSCs stemness also plays an important role. Summarizing traditional studies and we find many researchers ignored the influence of hypoxia on differentiated cells. We hypothesized that the residual differentiated cells may be dedifferentiated to GSC-like cells under hypoxia and play a crucial role in the rapid, high-frequency recurrence of GBM. Therefore, isolated CD133-CD15-NESTIN- cells were prepared as single-cell culture and treated with hypoxia. More than 95% of the surviving single differentiated CD133-CD15-NESTIN- cell dedifferentiated into tumorigenic CD133+CD15+NESTIN+ GSCs, and this process was regulated by hypoxia inducible factor-1α. Moreover, the serum also played an important role in this dedifferentiation. These findings challenge the traditional glioma cell heterogeneity model, cell division model and glioma malignancy development model. Our study also highlights the mechanism of GBM recurrence and the importance of anti-hypoxia therapy. In addition to GSCs, residual differentiated tumor cells also substantially contribute to treatment resistance and the rapid, high recurrence of GBM.
Collapse
|
16
|
Photodithazine photodynamic effect on viability of 9L/lacZ gliosarcoma cell line. Lasers Med Sci 2017; 32:1245-1252. [DOI: 10.1007/s10103-017-2227-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 04/28/2017] [Indexed: 10/19/2022]
|
17
|
van Straten D, Mashayekhi V, de Bruijn HS, Oliveira S, Robinson DJ. Oncologic Photodynamic Therapy: Basic Principles, Current Clinical Status and Future Directions. Cancers (Basel) 2017; 9:cancers9020019. [PMID: 28218708 PMCID: PMC5332942 DOI: 10.3390/cancers9020019] [Citation(s) in RCA: 618] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/10/2017] [Accepted: 02/12/2017] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) is a clinically approved cancer therapy, based on a photochemical reaction between a light activatable molecule or photosensitizer, light, and molecular oxygen. When these three harmless components are present together, reactive oxygen species are formed. These can directly damage cells and/or vasculature, and induce inflammatory and immune responses. PDT is a two-stage procedure, which starts with photosensitizer administration followed by a locally directed light exposure, with the aim of confined tumor destruction. Since its regulatory approval, over 30 years ago, PDT has been the subject of numerous studies and has proven to be an effective form of cancer therapy. This review provides an overview of the clinical trials conducted over the last 10 years, illustrating how PDT is applied in the clinic today. Furthermore, examples from ongoing clinical trials and the most recent preclinical studies are presented, to show the directions, in which PDT is headed, in the near and distant future. Despite the clinical success reported, PDT is still currently underutilized in the clinic. We also discuss the factors that hamper the exploration of this effective therapy and what should be changed to render it a more effective and more widely available option for patients.
Collapse
Affiliation(s)
- Demian van Straten
- Cell Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands.
| | - Vida Mashayekhi
- Cell Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands.
| | - Henriette S de Bruijn
- Center for Optical Diagnostics and Therapy, Department of Otolaryngology-Head and Neck Surgery, Erasmus Medical Center, Postbox 204, Rotterdam 3000 CA, The Netherlands.
| | - Sabrina Oliveira
- Cell Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands.
- Pharmaceutics, Department of Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht 3584 CG, The Netherlands.
| | - Dominic J Robinson
- Center for Optical Diagnostics and Therapy, Department of Otolaryngology-Head and Neck Surgery, Erasmus Medical Center, Postbox 204, Rotterdam 3000 CA, The Netherlands.
| |
Collapse
|
18
|
Fettweis G, Di Valentin E, L'homme L, Lassence C, Dequiedt F, Fillet M, Coupienne I, Piette J. RIP3 antagonizes a TSC2-mediated pro-survival pathway in glioblastoma cell death. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2017; 1864:113-124. [PMID: 27984090 DOI: 10.1016/j.bbamcr.2016.10.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/29/2016] [Accepted: 10/25/2016] [Indexed: 11/21/2022]
Abstract
Glioblastomas are the deadliest type of brain cancer and are frequently associated with poor prognosis and a high degree of recurrence despite removal by surgical resection and treatment by chemo- and radio-therapy. Photodynamic therapy (PDT) is a treatment well known to induce mainly necrotic and apoptotic cell death in solid tumors. 5-Aminolevulinic acid (5-ALA)-based PDT was recently shown to sensitize human glioblastoma cells (LN-18) to a RIP3 (Receptor Interacting Protein 3)-dependent cell death which is counter-acted by activation of autophagy. These promising results led us to investigate the pathways involved in cell death and survival mechanisms occurring in glioblastoma following PDT. In the present study, we describe a new TSC2 (Tuberous Sclerosis 2)-dependent survival pathway implicating MK2 (MAPKAPK2) kinase and 14-3-3 proteins which conducts to the activation of a pro-survival autophagy. Moreover, we characterized a new RIP3/TSC2 complex where RIP3 is suggested to promote cell death by targeting TSC2-dependent survival pathway. These results highlight (i) a new role of TSC2 to protect glioblastoma against PDT-induced cell death and (ii) TSC2 and 14-3-3 as new RIP3 partners.
Collapse
Affiliation(s)
- Gregory Fettweis
- Laboratory of Virology and Immunology, GIGA-I(3), University of Liège, Liège, Belgium
| | | | - Laurent L'homme
- Laboratory of Virology and Immunology, GIGA-I(3), University of Liège, Liège, Belgium
| | - Cédric Lassence
- Laboratory of Virology and Immunology, GIGA-I(3), University of Liège, Liège, Belgium
| | - Franck Dequiedt
- Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases, University of Liège, Liège, Belgium
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines (LAM), Department of Pharmaceutical Sciences, CIRM, University of Liège, Liège, Belgium
| | - Isabelle Coupienne
- Laboratory of Virology and Immunology, GIGA-I(3), University of Liège, Liège, Belgium
| | - Jacques Piette
- Laboratory of Virology and Immunology, GIGA-I(3), University of Liège, Liège, Belgium.
| |
Collapse
|
19
|
Lemaire L, Nel J, Franconi F, Bastiat G, Saulnier P. Perfluorocarbon-Loaded Lipid Nanocapsules to Assess the Dependence of U87-Human Glioblastoma Tumor pO2 on In Vitro Expansion Conditions. PLoS One 2016; 11:e0165479. [PMID: 27788227 PMCID: PMC5082913 DOI: 10.1371/journal.pone.0165479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 10/12/2016] [Indexed: 01/09/2023] Open
Abstract
Growing tumor cell lines, such as U87-MG glioma cells, under mild hypoxia (3% O2) leads to a ca. 40% reduction in growth rate once implanted in the brain of nude mice, as compared to normoxia (21% O2) grown cells, wherein the former over-express HIF-1 and VEGF-A. Despite developing differently, the tumors have similar: blood perfusion, oxygen consumption, and vascular surface area parameters, whereas the number of blood vessels is nearly doubled in the tumor arising from normoxia cultured cells. Interestingly, tumor oxygen tension, measured using 19F-oximetry, showed that the normoxia grown cells led to tumors characterized by mild hypoxic environment (approximately 4%) conditions, whilst the hypoxia grown cells led to tumors characterized by physioxic environment (approximately 6%) conditions. This reversal in oxygen concentration may be responsible for the apparent paradoxical growth profiles.
Collapse
Affiliation(s)
- Laurent Lemaire
- INSERM U 1066, 'Micro et Nanomédecines biomimétiques - MINT', Angers, France.,Université Angers, UMR-S1066, Angers, France
| | - Janske Nel
- INSERM U 1066, 'Micro et Nanomédecines biomimétiques - MINT', Angers, France.,Université Angers, UMR-S1066, Angers, France
| | | | - Guillaume Bastiat
- INSERM U 1066, 'Micro et Nanomédecines biomimétiques - MINT', Angers, France.,Université Angers, UMR-S1066, Angers, France
| | - Patrick Saulnier
- INSERM U 1066, 'Micro et Nanomédecines biomimétiques - MINT', Angers, France.,Université Angers, UMR-S1066, Angers, France
| |
Collapse
|
20
|
Schimanski A, Ebbert L, Sabel MC, Finocchiaro G, Lamszus K, Ewelt C, Etminan N, Fischer JC, Sorg RV. Human glioblastoma stem-like cells accumulate protoporphyrin IX when subjected to exogenous 5-aminolaevulinic acid, rendering them sensitive to photodynamic treatment. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 163:203-10. [DOI: 10.1016/j.jphotobiol.2016.08.043] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/25/2016] [Indexed: 01/01/2023]
|
21
|
Tong X, Srivatsan A, Jacobson O, Wang Y, Wang Z, Yang X, Niu G, Kiesewetter DO, Zheng H, Chen X. Monitoring Tumor Hypoxia Using (18)F-FMISO PET and Pharmacokinetics Modeling after Photodynamic Therapy. Sci Rep 2016; 6:31551. [PMID: 27546160 PMCID: PMC4992876 DOI: 10.1038/srep31551] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 07/13/2016] [Indexed: 11/09/2022] Open
Abstract
Photodynamic therapy (PDT) is an efficacious treatment for some types of cancers. However, PDT-induced tumor hypoxia as a result of oxygen consumption and vascular damage can reduce the efficacy of this therapy. Measuring and monitoring intrinsic and PDT-induced tumor hypoxia in vivo during PDT is of high interest for prognostic and treatment evaluation. In the present study, static and dynamic (18)F-FMISO PET were performed with mice bearing either U87MG or MDA-MB-435 tumor xenografts immediately before and after PDT at different time points. Significant difference in tumor hypoxia in response to PDT over time was found between the U87MG and MDA-MB-435 tumors in both static and dynamic PET. Dynamic PET with pharmacokinetics modeling further monitored the kinetics of (18)F-FMISO retention to hypoxic sites after treatment. The Ki and k3 parametric analysis provided information on tumor hypoxia by distinction of the specific tracer retention in hypoxic sites from its non-specific distribution in tumor. Dynamic (18)F-FMISO PET with pharmacokinetics modeling, complementary to static PET analysis, provides a potential imaging tool for more detailed and more accurate quantification of tumor hypoxia during PDT.
Collapse
Affiliation(s)
- Xiao Tong
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States.,Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Avinash Srivatsan
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Yu Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhantong Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Xiangyu Yang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Dale O Kiesewetter
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
22
|
Park KJ, Yu MO, Park DH, Park JY, Chung YG, Kang SH. Role of vincristine in the inhibition of angiogenesis in glioblastoma. Neurol Res 2016; 38:871-9. [PMID: 27472259 DOI: 10.1080/01616412.2016.1211231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Vincristine, a microtubule-destabilizing drug, was found to exhibit anti-angiogenic effects and anti-tumoral activity. However, the precise mechanism by which vincristine inhibits angiogenesis in glioblastomas is not well understood. Our aim was to investigate whether vincristine affects vascular endothelial growth factor (VEGF) expression in glioblastoma cells and determine whether it is mediated by the downregulation of hypoxia-inducible factor-1α (HIF-1α). METHODS We investigated the expression of HIF-1α in glioblastoma tissues resected from patients and in human glioblastoma cell lines using immunohistochemistry, Western blot analysis, and immunocytochemistry. In addition to an MTT assay assessing the effect of vincristine on cell proliferation and viability, the effects of vincristine on VEGF mRNA expression and HIF-1α protein were examined using real-time RT-PCR and Western blot analysis under 1% O2 (hypoxia). RESULTS HIF-1α was expressed in the majority of glioblastoma tissues and was detected mainly in the nucleus. Strong immunoreactivity for HIF- 1 α was found often in the hypercellular zones. Under hypoxic conditions, HIF-1α protein levels in the glioblastoma cell lines increased, primarily localizing into the nucleus similar to glioblastoma tissues. Exposure of glioblastoma cells to vincristine resulted in enrichment of the G2-M fraction of the cell cycle, which suggests that vincristine-mediated growth inhibition of glioblastoma is correlated with mitotic inhibition. Using doses lower than those found to reduce the viability and proliferation of cells by 50% (IC50), vincristine decreased both the expression of VEGF mRNA and the level of HIF-1α protein in hypoxic glioblastoma cells. In addition, following exposure to vincristine, the expression of VEGF mRNA was correlated with HIF-1α protein levels. CONCLUSIONS Our results suggest that the mechanism by which vincristine elicits an anti-angiogenic effect in glioblastomas under hypoxic conditions might be mediated, in part, by HIF-1α inhibition.
Collapse
Affiliation(s)
- Kyung-Jae Park
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Mi Ok Yu
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Dong-Hyuk Park
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Jung-Yul Park
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Yong-Gu Chung
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Shin-Hyuk Kang
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| |
Collapse
|
23
|
Kahlert UD, Mooney SM, Natsumeda M, Steiger HJ, Maciaczyk J. Targeting cancer stem-like cells in glioblastoma and colorectal cancer through metabolic pathways. Int J Cancer 2016; 140:10-22. [PMID: 27389307 DOI: 10.1002/ijc.30259] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/29/2016] [Accepted: 07/04/2016] [Indexed: 12/12/2022]
Abstract
Cancer stem-like cells (CSCs) are thought to be the main cause of tumor occurrence, progression and therapeutic resistance. Strong research efforts in the last decade have led to the development of several tailored approaches to target CSCs with some very promising clinical trials underway; however, until now no anti-CSC therapy has been approved for clinical use. Given the recent improvement in our understanding of how onco-proteins can manipulate cellular metabolic networks to promote tumorigenesis, cancer metabolism research may well lead to innovative strategies to identify novel regulators and downstream mediators of CSC maintenance. Interfering with distinct stages of CSC-associated metabolics may elucidate novel, more efficient strategies to target this highly malignant cell population. Here recent discoveries regarding the metabolic properties attributed to CSCs in glioblastoma (GBM) and malignant colorectal cancer (CRC) were summarized. The association between stem cell markers, the response to hypoxia and other environmental stresses including therapeutic insults as well as developmentally conserved signaling pathways with alterations in cellular bioenergetic networks were also discussed. The recent developments in metabolic imaging to identify CSCs were also summarized. This summary should comprehensively update basic and clinical scientists on the metabolic traits of CSCs in GBM and malignant CRC.
Collapse
Affiliation(s)
- U D Kahlert
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| | - S M Mooney
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - M Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - H-J Steiger
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| | - J Maciaczyk
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| |
Collapse
|
24
|
Xu CS, Wang ZF, Huang XD, Dai LM, Cao CJ, Li ZQ. Involvement of ROS-alpha v beta 3 integrin-FAK/Pyk2 in the inhibitory effect of melatonin on U251 glioma cell migration and invasion under hypoxia. J Transl Med 2015; 13:95. [PMID: 25889845 PMCID: PMC4371719 DOI: 10.1186/s12967-015-0454-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 03/06/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Melatonin, a well-known antioxidant, has been shown to possess anti-invasive properties for glioma. However, little is known about the effect of melatonin on glioma cell migration and invasion under hypoxia, which is a crucial microenvironment for tumor progress. In addition, focal adhesion kinase (FAK) and proline-rich tyrosine kinase 2 (Pyk2) are closely associated with cell migration and invasion. Therefore, we investigated the possible role of these kinases and its related signaling in the regulation of human U251 glioma cells behavior by melatonin under hypoxia. METHODS The abilities of migration and invasion of U251 glioma cells were determined by wound healing and transwell assay in vitro. The intracellular production of reactive oxygen species (ROS) was measured by using the fluorescent probe 6-carboxy-2', 7'-dichorodihydrofluorescein diacetate (DCFH-DA). Immunofluorescence experiments and western blotting analysis were used to detect the expression level of protein. Small interfering RNAs (siRNA) was used to silence specific gene expression. RESULTS The pharmacologic concentration (1 mM) of melatonin significantly inhibited the migration and invasion of human U251 glioma cells under hypoxia. The inhibitory effect of melatonin was accompanied with the reduced phosphorylation of FAK and Pyk2, and decreased expression of alpha v beta 3 (αvβ3) integrin. Additionally, inhibition of αvβ3 integrin by siRNA reduced the phosphorylation of FAK/Pyk2 and demonstrated the similar anti-tumor effects as melatonin, suggesting the involvement of αvβ3 integrin- FAK/Pyk2 pathway in the anti-migratory and anti-invasive effect of melatonin. It was also found that melatonin treatment decreased the ROS levels in U251 glioma cells cultured under hypoxia. ROS inhibitor apocynin not only inhibited αvβ3 integrin expression and the phosphorylation levels of FAK and Pyk2, but also suppressed the migratory and invasive capacity of U251 glioma cells under hypoxia. CONCLUSIONS These results suggest that melatonin exerts anti-migratory and anti-invasive effects on glioma cells in response to hypoxia via ROS-αvβ3 integrin-FAK/Pyk2 signaling pathways. This provides evidence that melatonin may be a potential therapeutic molecule targeting the hypoxic microenvironment of glioma.
Collapse
Affiliation(s)
- Cheng-Shi Xu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| | - Ze-Fen Wang
- Department of Physiology, School of basic medical science, Wuhan University, Wuhan, 430071, PR China.
| | - Xiao-Dong Huang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China. .,Department of Neurosurgery, Taihe Hospital of Shiyan, Shiyan, 442000, PR China.
| | - Li-Ming Dai
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| | - Chang-Jun Cao
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China. .,Laboratory of Neuro-oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| |
Collapse
|