1
|
Lisi L, Olivi A, Ciotti GMP, Marino S, Ferraro C, Menna G, Martire M, Pennisi G, Navarra P, Della Pepa GM. A topographic approach to the markers of macrophage/ microglia and other cell types in high grade glioma. Neurochem Int 2024:105922. [PMID: 39734023 DOI: 10.1016/j.neuint.2024.105922] [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: 09/06/2024] [Revised: 12/12/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024]
Abstract
In glioblastoma, glioma-associated microglia/macrophages (GAMs) represent the major population of tumor infiltrating cells, with up to one half of the cells of the tumor mass. Recent studies have shown that microglia are involved in the maintenance of immunological homeostasis and protection against autoimmunity. However, despite the growing body of evidence on the topic, many aspects are yet to be clarified. In our study, 3 different situations emerged concerning the markers of microglial/macrophage-related and other cell types in GBM patients: i) most of the markers (IBA1, TMEM119, CD206 and CD86) show an ascending gradient from the tumor center to the non-tumor/healthy area of the brain; ii) one marker (CD204) shows a descending gradient, going from the center of the tumor to the non-tumor/healthy brain area; iii) two markers (CD163 and P2RY12) show no gradient. These observations support the idea that the magnitude of the diverted inflammation is a 'extensive' rather than a 'local' phenomenon and that could possibly play a role in disease resistance and relapse.
Collapse
Affiliation(s)
- Lucia Lisi
- Dipartimento di Sicurezza e Bioetica, Università Cattolica del Sacro Cuore, IRCSS-Fondazione Policlinico Universitario Agostino Gemelli, 00168 Rome, Italy.
| | - Alessandro Olivi
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gabriella Maria Pia Ciotti
- Dipartimento di Sicurezza e Bioetica, Università Cattolica del Sacro Cuore, IRCSS-Fondazione Policlinico Universitario Agostino Gemelli, 00168 Rome, Italy
| | - Salvatore Marino
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Chiara Ferraro
- Dipartimento di Sicurezza e Bioetica, Università Cattolica del Sacro Cuore, IRCSS-Fondazione Policlinico Universitario Agostino Gemelli, 00168 Rome, Italy
| | - Grazia Menna
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Maria Martire
- Dipartimento di Sicurezza e Bioetica, Università Cattolica del Sacro Cuore, IRCSS-Fondazione Policlinico Universitario Agostino Gemelli, 00168 Rome, Italy
| | - Giovanni Pennisi
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Pierluigi Navarra
- Dipartimento di Sicurezza e Bioetica, Università Cattolica del Sacro Cuore, IRCSS-Fondazione Policlinico Universitario Agostino Gemelli, 00168 Rome, Italy
| | - Giuseppe Maria Della Pepa
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy.
| |
Collapse
|
2
|
Repetowski P, Warszyńska M, Dąbrowski JM. NIR-activated multifunctional agents for the combined application in cancer imaging and therapy. Adv Colloid Interface Sci 2024; 336:103356. [PMID: 39612723 DOI: 10.1016/j.cis.2024.103356] [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: 08/16/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024]
Abstract
Anticancer therapies that combine both diagnostic and therapeutic capabilities hold significant promise for enhancing treatment efficacy and patient outcomes. Among these, agents responsive to near-infrared (NIR) photons are of particular interest due to their negligible toxicity and multifunctionality. These compounds are not only effective in photodynamic therapy (PDT), but also serve as contrast agents in various imaging modalities, including fluorescence and photoacoustic imaging. In this review, we explore the photophysical and photochemical properties of NIR-activated porphyrin, cyanine, and phthalocyanines derivatives as well as aggregation-induced emission compounds, highlighting their application in synergistic detection, diagnosis, and therapy. Special attention is given to the design and optimization of these agents to achieve high photostability, efficient NIR absorption, and significant yields of fluorescence, heat, or reactive oxygen species (ROS) generation depending on the application. Additionally, we discuss the incorporation of these compounds into nanocarriers to enhance their solubility, stability, and target specificity. Such nanoparticle-based systems exhibit improved pharmacokinetics and pharmacodynamics, facilitating more effective tumor targeting and broadening the application range to photoacoustic imaging and photothermal therapy. Furthermore, we summarize the application of these NIR-responsive agents in multimodal imaging techniques, which combine the advantages of fluorescence and photoacoustic imaging to provide comprehensive diagnostic information. Finally, we address the current challenges and limitations of photodiagnosis and phototherapy and highlight some critical barriers to their clinical implementation. These include issues related to their phototoxicity, limited tissue penetration, and potential off-target effects. The review concludes by highlighting future research directions aimed at overcoming these obstacles, with a focus on the development of next-generation agents and platforms that offer enhanced therapeutic efficacy and imaging capabilities in the field of cancer treatment.
Collapse
Affiliation(s)
- Paweł Repetowski
- Faculty of Chemistry, Jagiellonian University, 30-387 Kraków, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Kraków, Poland
| | - Marta Warszyńska
- Faculty of Chemistry, Jagiellonian University, 30-387 Kraków, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Kraków, Poland
| | | |
Collapse
|
3
|
Pustimbara A, Umami RW, Prakoso NM, Rozaliyani A, Zaini J, Dwiranti A, Ogura SI, Bowolaksono A. Hemin Promotes Higher Effectiveness of Aminolevulinic-Photodynamic Therapy (ALA-PDT) in A549 Lung Cancer Cell Line by Interrupting ABCG2 Expression. Med Sci (Basel) 2024; 12:66. [PMID: 39584916 PMCID: PMC11587042 DOI: 10.3390/medsci12040066] [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: 10/17/2024] [Revised: 11/05/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND/OBJECTIVES Due to concerns about drug resistance and side effects, the discovery of improved drugs for lung cancer has attracted studies to find an effective and safe treatment. Aminolevulinic acid-mediated photodynamic therapy (ALA-PDT) is a cancer treatment with minimal side effects. However, ALA-PDT effectiveness can be hindered by ABCG2 and ABCB1 transporters impeding PpIX accumulation. Combining ALA with other substances can enhance PpIX accumulation. Hemin is a potential substance due to its antitumor properties and may be involved in regulating the ABCG2 and ABCB1 expressions. METHODS The objective of this report is to analyze the effects of administering a combination of hemin and ALA after 48 h on A549 lung cancer cells by quantifying cell viability, intracellular PpIX, and ROS accumulation, completed by ABCG2 and ABCB1 expressions. RESULTS Our data indicate that the combination of hemin and ALA followed by photoirradiation decreased the viability of A549 cells, which was due to increased intracellular PpIX and ROS. The expression of ABCG2 mRNA was significantly decreased after ALA-hemin treatment, while the ABCB1 mRNA expression increased. This result might suggest that ABCG2 plays a greater role than ABCB1 in regulating the PpIX accumulation in A549 lung cancer cells. CONCLUSIONS The combination of ALA and hemin followed by photoirradiation offers a promising novel treatment for lung cancer, and further evaluations of this therapy are required.
Collapse
Affiliation(s)
- Anantya Pustimbara
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok Campus, Depok 16424, Indonesia
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 B47, Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan;
| | - Rahma Wirdatul Umami
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok Campus, Depok 16424, Indonesia
| | - Nurul Muhammad Prakoso
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok Campus, Depok 16424, Indonesia
| | - Anna Rozaliyani
- Department of Parasitology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Jamal Zaini
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Indonesia, Persahabatan National Respiratory Referral Hospital, Jakarta 13230, Indonesia
| | - Astari Dwiranti
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok Campus, Depok 16424, Indonesia
| | - Shun-ichiro Ogura
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 B47, Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan;
| | - Anom Bowolaksono
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok Campus, Depok 16424, Indonesia
| |
Collapse
|
4
|
Ayyami Y, Ghorbani M, Dastgir M, Malekzadeh R, Mortezazadeh T. Chitosan-modified manganese oxide-conjugated methotrexate nanoparticles delivering 5-aminolevulinic acid as a dual-modal T1-T2* MRI contrast agent in U87MG cell detection. MAGMA (NEW YORK, N.Y.) 2024; 37:909-924. [PMID: 38795276 DOI: 10.1007/s10334-024-01169-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 05/27/2024]
Abstract
OBJECTIVE Glioblastoma multiforme is a highly aggressive form of brain cancer, and early diagnosis plays a pivotal role in improving patient survival rates. In this regard, molecular magnetic resonance imaging has emerged as a promising imaging modality due to its exceptional sensitivity to minute tissue changes and the ability to penetrate deep into the brain. This study aimed to assess the efficacy of a novel contrast agent in detecting gliomas during MRI scans. MATERIALS AND METHODS The contrast agent utilized modified chitosan coating on manganese oxide nanoparticles. The modification included adding methotrexate and 5-aminolevulinic acid (MnO2/CS@5-ALA-MTX) to target cells with overexpressed folate receptors and breaking down excess hydrogen peroxide in tumor tissue, resulting in enhanced signal intensity in T1-weighted MR images but diminished signal intensity in T2*-weighted MR images. RESULTS The nanosystem was characterized and evaluated in MR imaging, safety, and ability to target cells both in vivo and in vitro. MTX-free nanoparticles (MnO2/CS@5-ALA NPs) had no obvious cytotoxicity on cell lines U87MG and NIH3T3 after 24/48 h at a concentration of up to 160 µgr/mL (cell viability more than 80%). In this system, methotrexate enables tumor targeting and the MnO2/5-ALA improves T1-T2*-weighted MRI. In addition, MRI scans of mice with M109 carcinoma indicated significant tumor uptake and NP capacity to improve the positive contrast effect. CONCLUSION This developed MnO2/CS@5-ALA-MTX nanoparticle system may exhibit great potential in the accurate diagnosis of folate receptor over-expressing cancers such as glioblastoma.
Collapse
Affiliation(s)
- Yasin Ayyami
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marjan Ghorbani
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Iran Polymer and Petrochemical Institute, P.O.Box: 14965/115, Tehran, Iran
| | - Masoumeh Dastgir
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Malekzadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Tohid Mortezazadeh
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Goff NK, Ashby L, Ashour R. Meta-Analysis of the Efficacy of Raman Spectroscopy and Machine-Learning-Based Identification of Glioma Tissue. World Neurosurg 2024; 189:26-32. [PMID: 38796149 DOI: 10.1016/j.wneu.2024.05.112] [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: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 05/28/2024]
Abstract
Intraoperative Raman spectroscopy (RS) has been identified as a potential tool for surgeons to rapidly and noninvasively differentiate between diseased and normal tissue. Since the previous meta-analysis on the subject was published in 2016, improvements in both spectroscopy equipment and machine learning models used to process spectra may have led to an increase in RS efficacy. Therefore, we decided to conduct a meta-analysis to determine the efficacy of RS when differentiating between glioma tissue and normal brain tissue. Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed while conducting this meta-analysis. A search was conducted on PubMed and Web of Science for prospective and retrospective studies published between 2016 and 2022 using intraoperative RS and standard histology methods to differentiate between glioma and normal brain tissue. Meta-analyses of log odds ratios, sensitivity, and specificity were conducted in JASP using the random-effects model with restricted maximum likelihood estimation. A total of 9 studies met our inclusion criteria, comprising 673 patients and 8319 Raman spectra. Meta-analysis of log diagnostic odds ratios revealed high heterogeneity (I2 = 79.83%) and yielded a back-transformed diagnostic odds ratio of 76.71 (95% confidence interval: 39.57-148.71). Finally, meta-analysis for sensitivity and specificity of RS for glioma tissue showed high heterogeneity (I2 = 99.37% and 98.21%, respectively) and yielded an overall sensitivity of 95.3% (95% confidence interval: 91.0%-99.6%) and an overall specificity of 71.2% (95% confidence interval: 54.8%-87.6%). Calculation of a summary receiver operating curve yielded an overall area under the curve of 0.9265. Raman spectroscopy represents a promising tool for surgeons to quickly and accurately differentiate between healthy brain tissue and glioma tissue.
Collapse
Affiliation(s)
- Nicolas K Goff
- Department of Neurosurgery, The University of Texas at Austin Dell Medical School, Austin, Texas, USA.
| | - Landon Ashby
- Department of Neurosurgery, The University of Texas at Austin Dell Medical School, Austin, Texas, USA
| | - Ramsey Ashour
- Department of Neurosurgery, The University of Texas at Austin Dell Medical School, Austin, Texas, USA
| |
Collapse
|
6
|
Picart T, Gautheron A, Caredda C, Ray C, Mahieu-Williame L, Montcel B, Guyotat J. Fluorescence-Guided Surgical Techniques in Adult Diffuse Low-Grade Gliomas: State-of-the-Art and Emerging Techniques: A Systematic Review. Cancers (Basel) 2024; 16:2698. [PMID: 39123426 PMCID: PMC11311317 DOI: 10.3390/cancers16152698] [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: 06/28/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Diffuse low-grade gliomas are infiltrative tumors whose margins are not distinguishable from the adjacent healthy brain parenchyma. The aim was to precisely examine the results provided by the intraoperative use of macroscopic fluorescence in diffuse low-grade gliomas and to describe the new fluorescence-based techniques capable of guiding the resection of low-grade gliomas. Only about 20% and 50% of low-grade gliomas are macroscopically fluorescent after 5-amino-levulinic acid (5-ALA) or fluorescein sodium intake, respectively. However, 5-ALA is helpful for detecting anaplastic foci, and thus choosing the best biopsy targets in diffuse gliomas. Spectroscopic detection of 5-ALA-induced fluorescence can detect very low and non-macroscopically visible concentrations of protoporphyrin IX, a 5-ALA metabolite, and, consequently, has excellent performances for the detection of low-grade gliomas. Moreover, these tumors have a specific spectroscopic signature with two fluorescence emission peaks, which is useful for distinguishing them not only from healthy brain but also from high-grade gliomas. Confocal laser endomicroscopy can generate intraoperative optic biopsies, but its sensitivity remains limited. In the future, the coupled measurement of autofluorescence and induced fluorescence, and the introduction of fluorescence detection technologies providing a wider field of view could result in the development of operator-friendly tools implementable in the operative routine.
Collapse
Affiliation(s)
- Thiebaud Picart
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Groupe Hospitalier Est, Hospices Civils de Lyon, 59 Boulevard Pinel, 69500 Bron, France
- Faculty of Medicine Lyon Est, Université Claude Bernard Lyon 1, 8 Avenue Rockefeller, 69003 Lyon, France
- Cancer Research Centre of Lyon (CRCL) Inserm 1052, CNRS 5286, 28 Rue Laennec, 69008 Lyon, France
| | - Arthur Gautheron
- Laboratoire Hubert Curien UMR 5516, Institut d’Optique Graduate School, CNRS, Université Jean Monnet Saint-Etienne, 42023 Saint-Etienne, France;
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| | - Charly Caredda
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| | - Cédric Ray
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| | - Laurent Mahieu-Williame
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| | - Bruno Montcel
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| | - Jacques Guyotat
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Groupe Hospitalier Est, Hospices Civils de Lyon, 59 Boulevard Pinel, 69500 Bron, France
- Faculty of Medicine Lyon Est, Université Claude Bernard Lyon 1, 8 Avenue Rockefeller, 69003 Lyon, France
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| |
Collapse
|
7
|
Kumawat C, Takahashi T, Date I, Tomita Y, Tanaka M, Arataki S, Komatsubara T, Flores AOP, Yu D, Jain M. State-of-the-Art and New Treatment Approaches for Spinal Cord Tumors. Cancers (Basel) 2024; 16:2360. [PMID: 39001422 PMCID: PMC11240441 DOI: 10.3390/cancers16132360] [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: 05/21/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Spinal cord tumors, though rare, present formidable challenges in clinical management due to their intricate nature. Traditional treatment modalities like surgery, radiation therapy, and chemotherapy have been the mainstay for managing these tumors. However, despite significant advancements, challenges persist, including the limitations of surgical resection and the potential side effects associated with radiation therapy. In response to these limitations, a wave of innovative approaches is reshaping the treatment landscape for spinal cord tumors. Advancements in gene therapy, immunotherapy, and targeted therapy are offering groundbreaking possibilities. Gene therapy holds the potential to modify the genes responsible for tumor growth, while immunotherapy harnesses the body's own immune system to fight cancer cells. Targeted therapy aims to strike a specific vulnerability within the tumor cells, offering a more precise and potentially less toxic approach. Additionally, novel surgical adjuncts are being explored to improve visualization and minimize damage to surrounding healthy tissue during tumor removal. These developments pave the way for a future of personalized medicine for spinal cord tumors. By delving deeper into the molecular makeup of individual tumors, doctors can tailor treatment strategies to target specific mutations and vulnerabilities. This personalized approach offers the potential for more effective interventions with fewer side effects, ultimately leading to improved patient outcomes and a better quality of life. This evolving landscape of spinal cord tumor management signifies the crucial integration of established and innovative strategies to create a brighter future for patients battling this complex condition.
Collapse
Affiliation(s)
- Chetan Kumawat
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
- Department of Orthopedic Surgery, Sir Ganga Ram Hospital, Rajinder Nagar, New Delhi 110060, India
| | - Toshiyuki Takahashi
- Spinal Disorder Center, Fujieda Heisei Memorial Hospital, 123-1 Mizuue Fujieda, Shizuoka 426-8662, Japan
| | - Isao Date
- Department of Neurosurgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Yousuke Tomita
- Department of Neurosurgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Masato Tanaka
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Shinya Arataki
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Tadashi Komatsubara
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Angel O P Flores
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Dongwoo Yu
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Mukul Jain
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| |
Collapse
|
8
|
Oudin A, Moreno-Sanchez PM, Baus V, Niclou SP, Golebiewska A. Magnetic resonance imaging-guided intracranial resection of glioblastoma tumors in patient-derived orthotopic xenografts leads to clinically relevant tumor recurrence. BMC Cancer 2024; 24:3. [PMID: 38166949 PMCID: PMC10763155 DOI: 10.1186/s12885-023-11774-6] [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: 07/07/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Preclinical in vivo cancer models are essential tools for investigating tumor progression and response to treatment prior to clinical trials. Although treatment modalities are regularly assessed in mice upon tumor growth in vivo, surgical resection remains challenging, particularly in the orthotopic site. Here, we report a successful surgical resection of glioblastoma (GBM) in patient-derived orthotopic xenografts (PDOXs). METHODS We derived a cohort of 46 GBM PDOX models that faithfully recapitulate human disease in mice. We assessed the detection and quantification of intracranial tumors using magnetic resonance imaging (MRI).To evaluate feasibility of surgical resection in PDOXs, we selected two models representing histopathological features of GBM tumors, including diffuse growth into the mouse brain. Surgical resection in the mouse brains was performed based on MRI-guided coordinates. Survival study followed by MRI and immunohistochemistry-based evaluation of recurrent tumors allowed for assessment of clinically relevant parameters. RESULTS We demonstrate the utility of MRI for the noninvasive assessment of in vivo tumor growth, preoperative programming of resection coordinates and follow-up of tumor recurrence. We report tumor detection by MRI in 90% of GBM PDOX models (36/40), of which 55% (22/40) can be reliably quantified during tumor growth. We show that a surgical resection protocol in mice carrying diffuse primary GBM tumors in the brain leads to clinically relevant outcomes. Similar to neurosurgery in patients, we achieved a near total to complete extent of tumor resection, and mice with resected tumors presented significantly increased survival. The remaining unresected GBM cells that invaded the normal mouse brain prior to surgery regrew tumors with similar histopathological features and tumor microenvironments to the primary tumors. CONCLUSIONS Our data positions GBM PDOXs developed in mouse brains as a valuable preclinical model for conducting therapeutic studies that involve surgical tumor resection. The high detectability of tumors by MRI across a substantial number of PDOX models in mice will allow for scalability of our approach toward specific tumor types for efficacy studies in precision medicine-oriented approaches. Additionally, these models hold promise for the development of enhanced image-guided surgery protocols.
Collapse
Affiliation(s)
- Anais Oudin
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), 6A, Rue Nicolas-Ernest Barblé, Luxembourg, L-1210, Luxembourg
| | - Pilar M Moreno-Sanchez
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), 6A, Rue Nicolas-Ernest Barblé, Luxembourg, L-1210, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Belvaux, L-4367, Luxembourg
| | - Virginie Baus
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), 6A, Rue Nicolas-Ernest Barblé, Luxembourg, L-1210, Luxembourg
| | - Simone P Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), 6A, Rue Nicolas-Ernest Barblé, Luxembourg, L-1210, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Belvaux, L-4367, Luxembourg
| | - Anna Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), 6A, Rue Nicolas-Ernest Barblé, Luxembourg, L-1210, Luxembourg.
| |
Collapse
|
9
|
Sansaloni-Pastor S, Lange N. Unleashing the potential of 5-Aminolevulinic acid: Unveiling a promising target for cancer diagnosis and treatment beyond photodynamic therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 247:112771. [PMID: 37647818 DOI: 10.1016/j.jphotobiol.2023.112771] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
The therapeutic properties of 5-aminolevulinic acid (5-ALA) have been extensively studied for cancer detection and treatment using photodynamic therapy (PDT). When administered externally, 5-ALA is converted to protoporphyrin IX (PpIX) in cancer cells, which generates reactive oxygen species (ROS) upon exposure to light. This process enables targeted cell death induction and cancer detection. Given the highly conserved nature of heme biosynthesis over billions of years, we hypothesized that natural mechanisms might exist to prevent excessive accumulation of PpIX or heme resulting from 5-ALA overload. Therefore, we anticipated alterations in protein expression profiles upon exogenous administration of 5-ALA. To understand cellular responses to 5-ALA, we investigated protein expression changes and identified OR1B1 as a promising target in bladder, prostate, lung, and cervical cancer cells. OR1B1 expression was observed only with 5-ALA and ferrous chloride, highlighting the central role of heme in this discovery. Immunofluorescence and electron microscopy confirmed OR1B1's sub-cellular localization. These findings suggest that 5-ALA transformation in cancer cells and OR1B1 expression have potential for enhancing cancer detection and developing alternative treatments, including immunotherapy. This approach overcomes the limitations of PDT and opens new avenues for effective and targeted cancer interventions.
Collapse
Affiliation(s)
- Sara Sansaloni-Pastor
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland
| | - Norbert Lange
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland.
| |
Collapse
|
10
|
García-Montaño LA, Licón-Muñoz Y, Martinez FJ, Keddari YR, Ziemke MK, Chohan MO, Piccirillo SG. Dissecting Intra-tumor Heterogeneity in the Glioblastoma Microenvironment Using Fluorescence-Guided Multiple Sampling. Mol Cancer Res 2023; 21:755-767. [PMID: 37255362 PMCID: PMC10390891 DOI: 10.1158/1541-7786.mcr-23-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/25/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
The treatment of the most aggressive primary brain tumor in adults, glioblastoma (GBM), is challenging due to its heterogeneous nature, invasive potential, and poor response to chemo- and radiotherapy. As a result, GBM inevitably recurs and only a few patients survive 5 years post-diagnosis. GBM is characterized by extensive phenotypic and genetic heterogeneity, creating a diversified genetic landscape and a network of biological interactions between subclones, ultimately promoting tumor growth and therapeutic resistance. This includes spatial and temporal changes in the tumor microenvironment, which influence cellular and molecular programs in GBM and therapeutic responses. However, dissecting phenotypic and genetic heterogeneity at spatial and temporal levels is extremely challenging, and the dynamics of the GBM microenvironment cannot be captured by analysis of a single tumor sample. In this review, we discuss the current research on GBM heterogeneity, in particular, the utility and potential applications of fluorescence-guided multiple sampling to dissect phenotypic and genetic intra-tumor heterogeneity in the GBM microenvironment, identify tumor and non-tumor cell interactions and novel therapeutic targets in areas that are key for tumor growth and recurrence, and improve the molecular classification of GBM.
Collapse
Affiliation(s)
- Leopoldo A. García-Montaño
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Yamhilette Licón-Muñoz
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Frank J. Martinez
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Yasine R. Keddari
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of California, Merced, California
| | - Michael K. Ziemke
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Muhammad O. Chohan
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sara G.M. Piccirillo
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| |
Collapse
|
11
|
Cheng M, Hu C, Yao Z, Hao D, Jin T, Zhang Z, Liu X, Yu Z, Zhang H. Harnessing Reconstructed Macrophage Modulation of Infiltration-Excluded Immune Microenvironments To Delineate Glioma Infiltrative Region. ACS APPLIED MATERIALS & INTERFACES 2023; 15:8811-8823. [PMID: 36758126 DOI: 10.1021/acsami.2c16586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
High invasiveness of glioma produces residual glioma cells in the brain parenchyma after surgery and ultimately causes recurrence. Precise delineation of glioma infiltrative region is critical for an accurate complete resection, which is challenging. The glioma-infiltrating area constitutes infiltration-excluded immune microenvironments (I-E TIMEs), which recruits endogenous or adoptive macrophages to the invasive edge of glioma. Thus, combined with immune cell tracing technology, we provided a novel strategy for the preoperative precise definition of the glioma infiltration boundary, even satellite-like infiltration stoves. Herein, the biomimetic probe was constructed by internalizing fluorophore labeled PEGylated KMnF3 nanoparticles into bone-marrow-derived macrophages using magnetic resonance imaging (MRI)/fluorescence imaging (FI). The biomimetic probe was able to cross the blood-brain barrier and home to the orthotopic glioma infiltrates including satellite stove under MRI and FI tracing, which was validated using hematoxylin and eosin staining, indicating its excellent performance in distinguishing the margins between the glioma cell and normal tissues. This study guides the precise definition of glioma infiltration boundaries at the cellular level, including the observation of any residual glioma cells after surgery. Thus, it has the potential to guide surgery to maximize resection and predict recurrence in the clinic.
Collapse
Affiliation(s)
- Miaomiao Cheng
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chenchen Hu
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao 266071, China
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Dapeng Hao
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Teng Jin
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430020, China
| | - Zhenao Zhang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xuejun Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Zhengze Yu
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao 266071, China
| | - Hua Zhang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|