留言板

尊敬的读者、作者、审稿人, 关于本刊的投稿、审稿、编辑和出版的任何问题, 您可以本页添加留言。我们将尽快给您答复。谢谢您的支持!

姓名
邮箱
手机号码
标题
留言内容
验证码

拉曼光谱技术在脑胶质瘤检测中的应用研究

葛梅兰 王与烨 李海滨 徐德刚 姚建铨

葛梅兰, 王与烨, 李海滨, 徐德刚, 姚建铨. 拉曼光谱技术在脑胶质瘤检测中的应用研究[J]. 中国光学(中英文). doi: 10.37188/CO.2024-0003
引用本文: 葛梅兰, 王与烨, 李海滨, 徐德刚, 姚建铨. 拉曼光谱技术在脑胶质瘤检测中的应用研究[J]. 中国光学(中英文). doi: 10.37188/CO.2024-0003
GE Mei-lan, WANG Yu-ye, LI Hai-bin, XU De-gang, YAO Jian-quan. Application of Raman spectroscopy in the detection of brain glioma[J]. Chinese Optics. doi: 10.37188/CO.2024-0003
Citation: GE Mei-lan, WANG Yu-ye, LI Hai-bin, XU De-gang, YAO Jian-quan. Application of Raman spectroscopy in the detection of brain glioma[J]. Chinese Optics. doi: 10.37188/CO.2024-0003

拉曼光谱技术在脑胶质瘤检测中的应用研究

doi: 10.37188/CO.2024-0003
基金项目: 国家自然科学基金(No. 62175182,U22A20353,62275193,U22A20123)
详细信息
    作者简介:

    葛梅兰(1991—),女,陕西宝鸡人,博士,主要研究方向为太赫兹-拉曼光谱技术及其在生物医学中的应用研究。E-mail:meilange@tju.edu.cn

    王与烨(1983—),女,山西朔州人,教授,博士生导师,2009年于天津大学获得博士学位,主要研究方向为太赫兹光子学辐射源技术及其应用。E-mail:yuyewang@tju.edu.cn

    李海滨(1998—),女,河北保定人,博士研究生在读,2020年于长春理工大学获得学士学位,主要研究方向为太赫兹生物医学成像研究。E-mail:haibin_li@tju.edu.cn

    徐德刚(1974—),男,山东青岛人,教授,博士生导师,2005年于天津大学获得博士学位,主要研究方向为激光与光电子技术、太赫兹技术及激光雷达等研究工作。E-mail:xudegangtju.@edu.cn

    姚建铨(1939—),男,上海市人,中国科学院院士。1957年毕业于天津大学精仪系,著名激光与非线性光学专家。E-mail: jqyao@tju.edu.cn

  • 中图分类号: TN29

Application of Raman spectroscopy in the detection of brain glioma

Funds: Supported by National Natural Science Foundation of China (No. U22A20353, 62175182, 62275193, U22A20123)
More Information
  • 摘要:

    脑胶质瘤是一种常见的颅内原发性肿瘤,具有治愈率、复发率高等特点。脑胶质瘤边界的精准识别是减少患者术后复发、改善愈后的重要前提。因此,发展快速、高灵敏度、无标记的脑胶质瘤诊断方法具有重要的临床意义。拉曼光谱技术作为一种指纹谱,能够在分子水平上反映物质的化学和结构信息,已经在脑胶质瘤的定性定位识别中表现出巨大的应用前景。本文首先介绍了不同种类的拉曼光谱技术,其次梳理了拉曼光谱技术在脑胶质瘤中的研究现状,最后对拉曼光谱技术在脑胶质瘤检测中的未来发展进行展望。

     

  • 图 1  Raman光谱在脑胶质瘤检测中的研究现状

    Figure 1.  The research state of the detection of brain glioma through Raman spectroscopy

    图 2  A和B为未染色人胶质母细胞瘤冰冻切片在显微镜下的照片;C和D为组织切片的Raman光谱绘制的伪彩色图(对应于A和B)红色:主要的肿瘤组织区域,蓝色:坏死组织;E和F为H&E染色后照片;G为F图中绿框处放大后的图片[34]

    Figure 2.  A and B, Photomicrographs of unstained human glioblastoma cryosections used in Raman mapping experiments; C and D, Pseudo-color Raman maps of Raman spectra of the tissue sections shown in A and B; Red: areas of vital tumor tissue; blue: areas of necrosis; yellow: areas in the scan where no tissue was present (edges, freezing artifacts); E and F, Photomicrographs of the same tissue sections after H&E staining; G, ×40 magnification of detail marked with a green frame in F[34]

    图 3  浸润性组织的(1)白质、(2)出血和(3)高密度细胞肿瘤区域的Raman光谱[35]

    Figure 3.  Raman spectra (left) from regions identified as (1) infiltrating grey matter, (2) hemorrhage, and (3) hypercellular tumor from tissue; (right) Raman imaging result of different components in infiltration tissue[35]

    图 4  IDH1突变型(IDH1-mut)和野生型(IDH1-wt)的Raman光谱对比[38]

    Figure 4.  Comparison of Raman spectra of IDH1-wt and IDH1-mut glioma[38]

    图 5  健康(蓝色)和胶质瘤组织(红色)的平均Raman光谱[39]

    Figure 5.  Normalized mean spectra with standard deviation for healthy (blue) and tumor patients (red)[39]

    图 6  (a)拉曼检测系统示意图;(b)小鼠用于在体拉曼检测的图片[40]

    Figure 6.  (a) Schematic setup of the Raman system; (b) Image of Raman measurement based on mouse model in vivo [40]

    图 7  (a)皮层暴露后大鼠脑组织;(b)Raman光谱得到伪彩色图,其中蓝色、青色和黄色为正常组织,红色为血管,灰色为肿瘤组织,黑色为肿瘤边缘;(c)组织的显微照片和Raman图像的叠加[40]

    Figure 7.  (a) Deceased mouse with exposed cortex; (b) Raman images are segmented by cluster analysis: Normal brain tissue is depicted in blue, cyan, and yellow, the red is blood vessel, and the tumor and tumor margin are shown in gray and black, respectively; (c) Overlay of the photomicrograph and the Raman image [40]

    图 8  手持接触式Raman光谱探针,(a)实验系统示意图;(b)基于探针的脑组织检测图[41]

    Figure 8.  The handheld contact fiber optic probe for Raman spectroscopy, (a) Experimental setup diagram; (b) The probe is used to interrogate brain tissue during surgery[41]

    图 9  (a)商用活检针内部的拉曼微探针组件的分解图;(b)拉曼检测时神经导航系统的图像[43]

    Figure 9.  (a) Exploded view of the Raman microprobe components alongside the internal cannula of the commercial brain biopsy needle; (b) Images from the neuronavigation system[43]

    图 10  (a)胶质瘤切除术期间,使用手持式接触探针在致密肿瘤组织(红色)、浸润性组织(黄色)和周围正常组织位置图;(b)在体致密性肿瘤组织、浸润性组织和正常组织的高波数Raman光谱,(c)每种类型组织的H&E染色显微图片[44]

    Figure 10.  (a) Schematic depiction of in vivo Raman spectral measurements taken in the surgical cavity during glioma resection using the handheld contact probe in dense cancer (red), infltrated brain (yellow) and surrounding normal brain; (b) In vivo high wavenumber Raman spectra of dense cancer, infltrated brain and normal brain, averaged over all samples; (c) Representative H&E-stained micrographs for each tissue type[44]

    图 11  (a)脑胶质瘤患者中,正常脑组织、浸润脑组织和致密型肿瘤脑组织中脂质和蛋白的Raman强度比值(2930 cm−1/2845 cm−1)的箱线图,(b)基于SVM算法的受试者特性曲线[44]

    Figure 11.  (a) Boxplots of the ratio of the lipid and protein bands (2930 cm−1 /2845 cm−1) for normal brain, infltrated brain and dense cancer tissue in glioma patients; (b) Receiver operating characteristic curve computed using the SVM algorithm[44]

    图 12  正常组织和不同恶性胶质瘤组织中(a)1588/1440cm−1和(b)2934/2885cm−1 Raman强度比值,其中G0-N为正常脑组织;GI:I级胶质瘤;GII:II级胶质瘤;GIII:III级胶质瘤;GIV:IV级胶质瘤[45]

    Figure 12.  The ratios of (a) I1588 cm−1 ∕I1440 cm−1 and (b) I2934 cm−1 ∕I2885 cm−1 from normal human brain tissues and glioma tissues with increasing malignancy; G0-N: normal human brain tissues; GI: grade I; GII: grade II; GIII: grade III; GIV: grade IV[45].

    图 13  基于正常、低级别胶质瘤样本与高级别胶质瘤样本的Raman光谱,主成分PC2和PC1的聚类结果[45]

    Figure 13.  The clustering result of principal components PC1 and PC2 based on the Raman spectra of normal tissue, low grade (I and II) , and high grade (III and IV) glioma[45]

    图 14  健康小鼠大脑的CARS显微图像[46]

    Figure 14.  CARS microscopy image of a healthy mouse brain[46]

    图 15  (a)为图1-14中白色框中的放大图像;(b)为(a)图中对应位置处的H&E图像[46]

    Figure 15.  (a) The enlarged image from the white rectangle in the Fig. 14; (b) H&E image of the same region of the same mouse brain[46]

    图 16  小鼠脑中星形细胞瘤的CARS图像,(a)低分辨率、大视场的CARS显微镜图像;(b)为(a)图中矩形区域中局部组织的CARS图像[46]

    Figure 16.  CARS images of astrocytoma in mouse, (a) Alow resolution, large field of view mosaic CARS microscopy image; (b) The CARS image of local tissue from the rectangle in (a) [46]

    图 17  (a)小鼠脑中胶质母细胞瘤的可见光图像,其中黑色线为肿瘤边界,青色为感兴趣区域;(b)特定区域内近距离显微图像;(c)肿瘤和正常脑组织的伪彩色图像,蓝色为细胞核、红色为脂质含量、绿色为红细胞;(d)利用细胞核(蓝色)、脂质含量(红色)绘制的CARS图像;(e)细胞核(蓝色)、脂质含量(红色)、CH3-CH2拉伸(绿色)绘制的CARS图像,NB为正常脑组织,T为肿瘤细胞,WM为白质;(f)肿瘤、白质和正常组织的CARS光谱[47]

    Figure 17.  (a) Brightfield image of xenograft glioblastoma in mouse brain, with the tumor hard boundary outlined (black). The cyan indicates a region of interest (ROI); (b) Phase contrast micrograph of CARS ROIs; (c) Pseudocolour CARS image of tumor and normal brain tissue, with nuclei highlighted in blue, lipid content in red and red blood cells in green; (d) CARS image and axial scan with nuclei highlighted in blue and lipid content in red; (e) CARS image with nuclei highlighted in blue, lipid content in red and CH3 stretch–CH2 stretch in green, NB: normal brain; T: tumor cells, WM: white matter; (f) Normalized CARS of different tissue[47]

    图 18  新鲜小鼠组织的SRS图像,A特征峰2845cm−1处小鼠脑胼胝体神经元束中标记出髓鞘中含丰富CH2;B约1 mm厚脑组织切片CH2的SRS图像;C三个独立区域在同一深度处,小鼠耳皮肤中CH2的SRS图像。从左到右依次为:角质层、皮脂腺和皮下脂肪层;D角质层有无CH2共振的SRS和CARS图像对比。与CARS图像不同,SRS中没有非谐振背景[47]

    Figure 18.  SRS imaging of fresh mouse tissue, (A) Neuron bundles in corpus callosum of mouse brain imaged at 2845 cm−1 highlighting myelin sheaths rich in CH2; (B) SRS image of CH2 acquired from ~1 mm brain tissue; (C) SRS images of CH2 of mouse ear skin in the same depths. From left to right: stratum corneum, sebaceous gland, and subcutaneous fat layer; (D) Comparison of SRS and CARS images of stratum corneum on and off the CH2 resonance. Unlike CARS, SRL has no nonresonant background[47]

    图 19  (A)健康、(B)II级、(C)III级、(D)IV级胶质瘤组织不同位置的SERS光谱[50]

    Figure 19.  SERS spectra of (A) Healthily, (B) II grade, (C) III grade, (D) IV grade tissue[50]

    图 20  (a)神经胶质瘤细胞和正常星形胶质细胞中Raman光谱的测量;(b)神经胶质瘤细胞(红色)和正常星形胶质细胞(蓝色)Raman光谱之间的差异[50]

    Figure 20.  (a) Measurement of Raman scattering spectra in glioma cells and normal astrocytes; (b) The difference in Raman spectra between glioma cells (red) and normal astrocytes (blue)[50]

    图 21  C6和SK-N-SH细胞系分化、未分化和正常神经细胞的Raman光谱[52]

    Figure 21.  Raman spectra of undifferentiated and RA-differentiated C6 glioma and SK-N-SH cells in comparison with spectra of normal neural stem cells (NSCs) [52]

    图 22  分化(红色)和未分化(黑色)细胞的Raman特征峰的强度差异[52]

    Figure 22.  Difference in marker peak intensities for differentiated and undifferentiated cells: (a) C6 cells and (b) SK-N-SH cells[52]

    表  1  CARS、SRS和SERS三种非线性拉曼光谱技术的优缺点对比

    Table  1.   Comparison of advantages and disadvantages of the three nonlinear Raman spectroscopy techniques: CARS, SRS and SERS

    拉曼光谱
    技术
    CARSSRSSERS
    优点无标记无标记、
    标准谱
    克服荧光背景噪声
    缺点易受非共振背景噪声影响、非标准谱、系统复杂系统复杂、分子选择性强引入新的金属材料、基底制备工艺复杂
    下载: 导出CSV

    表  2  不同拉曼光谱技术在脑胶质瘤生物标志物中的应用

    Table  2.   Application of different Raman spectroscopy in the detection of brain glioma biomarkers

    年份拉曼技术标志物光谱范围/cm−1参考文献
    2021表面增强拉曼循环肿瘤DNA100-1800[71]
    2023表面增强拉曼血管内皮生成因子200-1800[54]
    2018共振拉曼光谱乳酸和三磷脂酸腺苷500-4000[72]
    2023共聚焦拉曼光谱糖基化400-1800[73]
    2023自发拉曼光谱γ-氨基丁酸0-150[74]
    下载: 导出CSV
  • [1] AGUIAR R P, FALCÃO E T, PASQUALUCCI C A, et al. Use of Raman spectroscopy to evaluate the biochemical composition of normal and tumoral human brain tissues for diagnosis[J]. Lasers in Medical Science, 2022, 37(1): 121-133. doi: 10.1007/s10103-020-03173-1
    [2] SCHUPPER A J, YONG R L, HADJIPANAYIS C G. The neurosurgeon's armamentarium for gliomas: an update on intraoperative technologies to improve extent of resection[J]. Journal of Clinical Medicine, 2021, 10(2): 236. doi: 10.3390/jcm10020236
    [3] GHINDA D C, WU J S, DUNCAN N W, et al. How much is enough-can resting state fMRI provide a demarcation for neurosurgical resection in glioma?[J]. Neuroscience & Biobehavioral Reviews, 2018, 84: 245-261.
    [4] WU L M, XU D G, WANG Y Y, et al. Study of in vivo brain glioma in a mouse model using continuous-wave terahertz reflection imaging[J]. Biomedical Optics Express, 2019, 10(8): 3953-3962. doi: 10.1364/BOE.10.003953
    [5] CHERKASOVA O, PENG Y, KONNIKOVA M, et al. Diagnosis of glioma molecular markers by terahertz technologies[J]. Photonics, 2021, 8(1): 22. doi: 10.3390/photonics8010022
    [6] ZHANG Y, YU H Q, LI Y Q, et al. Raman spectroscopy: a prospective intraoperative visualization technique for gliomas[J]. Frontiers in Oncology, 2023, 12: 1086643. doi: 10.3389/fonc.2022.1086643
    [7] YOO W S, KIM J H, HAN S M. Multiwavelength Raman characterization of silicon stress near through-silicon vias and its inline monitoring applications[J]. Journal of Micro/Nanolithography, MEMS, and MOEMS, 2014, 13(1): 011205. doi: 10.1117/1.JMM.13.1.011205
    [8] SEKAR S K V, MOSCA S, FARINA A, et al. Frequency offset Raman spectroscopy (FORS) for depth probing of diffusive media[J]. Optics Express, 2017, 25(5): 4585-4597. doi: 10.1364/OE.25.004585
    [9] PUPPELS G J, DE MUL F F M, OTTO C, et al. Studying single living cells and chromosomes by confocal Raman microspectroscopy[J]. Nature, 1990, 347(6290): 301-303. doi: 10.1038/347301a0
    [10] CUI S SH, ZHANG SH, YUE SH H. Raman spectroscopy and imaging for cancer diagnosis[J]. Journal of Healthcare Engineering, 2018, 2018: 8619342.
    [11] RAUWEL E, AL-ARAG S, SALEHI H, et al. Assessing cobalt metal nanoparticles uptake by cancer cells using live Raman spectroscopy[J]. International Journal of Nanomedicine, 2020, 15: 7051-7062. doi: 10.2147/IJN.S258060
    [12] ROBERT B. Resonance Raman spectroscopy[J]. Photosynthesis Research, 2009, 101(2-3): 147-155. doi: 10.1007/s11120-009-9440-4
    [13] HOWARD W F, NELSON W H, SPERRY J F. A resonance Raman method for the rapid detection and identification of bacteria in water[J]. Applied Spectroscopy, 1980, 34(1): 72-75. doi: 10.1366/0003702804730790
    [14] HAN Y K, QIANG L, GAO Y K, et al. Large-area surface-enhanced Raman spectroscopy substrate by hybrid porous GaN with Au/Ag for breast cancer miRNA detection[J]. Applied Surface Science, 2021, 541: 148456. doi: 10.1016/j.apsusc.2020.148456
    [15] ZHANG J, DONG Y H, ZHU W F, et al. Ultrasensitive detection of circulating tumor DNA of lung cancer via an enzymatically amplified SERS-based frequency shift assay[J]. ACS Applied Materials & Interfaces, 2019, 11(20): 18145-18152.
    [16] ANDREI C C, MORAILLON A, LARQUET E, et al. SERS characterization of aggregated and isolated bacteria deposited on silver-based substrates[J]. Analytical and Bioanalytical Chemistry, 2021, 413(5): 1417-1428. doi: 10.1007/s00216-020-03106-5
    [17] CHEN H, DAS A, BI L Y, et al. Recent advances in surface-enhanced Raman scattering-based microdevices for point-of-care diagnosis of viruses and bacteria[J]. Nanoscale, 2020, 12(42): 21560-21570. doi: 10.1039/D0NR06340A
    [18] JAWORSKA A, FORNASARO S, SERGO V, et al. Potential of surface enhanced Raman spectroscopy (SERS) in therapeutic drug monitoring (TDM). A critical review[J]. Biosensors, 2016, 6(3): 47. doi: 10.3390/bios6030047
    [19] HUNTER R, SOHI A N, KHATOON Z, et al. Optofluidic label-free SERS platform for rapid bacteria detection in serum[J]. Sensors and Actuators B: Chemical, 2019, 300: 126907. doi: 10.1016/j.snb.2019.126907
    [20] ZHANG B Y, YIN P J, HU Y H, et al. Highly accurate and label-free discrimination of single cancer cell using a plasmonic oxide-based nanoprobe[J]. Biosensors and Bioelectronics, 2022, 198: 113814. doi: 10.1016/j.bios.2021.113814
    [21] DEY P, VAIDEANU A, MOSCA S, et al. Surface enhanced deep Raman detection of cancer tumour through 71 mm of heterogeneous tissue[J]. Nanotheranostics, 2022, 6(3): 337-349. doi: 10.7150/ntno.71510
    [22] POTMA E O, DE BOEIJ W P, VAN HAASTERT P J M, et al. Real-time visualization of intracellular hydrodynamics in single living cells[J]. Proceedings of the National Academy of Sciences of the United States of America, 2001, 98(4): 1577-1582.
    [23] STEUWE C, PATEL I I, UL-HASAN M, et al. CARS based label-free assay for assessment of drugs by monitoring lipid droplets in tumour cells[J]. Journal of Biophotonics, 2014, 7(11-12): 906-913. doi: 10.1002/jbio.201300110
    [24] WANG H W, LANGOHR I M, STUREK M, et al. Imaging and quantitative analysis of atherosclerotic lesions by CARS-based multimodal nonlinear optical microscopy[J]. Arteriosclerosis, Thrombosis, and Vascular Biology, 2009, 29(9): 1342-1348. doi: 10.1161/ATVBAHA.109.189316
    [25] KISS N, KROLOPP Á, LŐRINCZ K, et al. Stain-free histopathology of basal cell carcinoma by dual vibration resonance frequency CARS microscopy[J]. Pathology & Oncology Research, 2018, 24(4): 927-930.
    [26] WEI L, YU Y, SHEN Y H, et al. Vibrational imaging of newly synthesized proteins in live cells by stimulated Raman scattering microscopy[J]. Proceedings of the National Academy of Sciences of the United States of America, 2013, 110(28): 11226-11231.
    [27] OZEKI Y, UMEMURA W, OTSUKA Y, et al. High-speed molecular spectral imaging of tissue with stimulated Raman scattering[J]. Nature Photonics, 2012, 6(12): 845-851. doi: 10.1038/nphoton.2012.263
    [28] SAAR B G, FREUDIGER C W, REICHMAN J, et al. Video-rate molecular imaging in vivo with stimulated Raman scattering[J]. Science, 2010, 330(6009): 1368-1370. doi: 10.1126/science.1197236
    [29] FU D, LU F K, ZHANG X, et al. Quantitative chemical imaging with multiplex stimulated Raman scattering microscopy[J]. Journal of the American Chemical Society, 2012, 134(8): 3623-3626. doi: 10.1021/ja210081h
    [30] ZHANG R R, KUO J S. Detection of human brain tumor infiltration with quantitative stimulated Raman scattering microscopy[J]. Neurosurgery, 2016, 78(4): N9-N11.
    [31] LU F K, CALLIGARIS D, OLUBIYI O I, et al. Label-free neurosurgical pathology with stimulated Raman imaging[J]. Cancer Research, 2016, 76(12): 3451-3462. doi: 10.1158/0008-5472.CAN-16-0270
    [32] REINECKE D, VON SPRECKELSEN N, MAWRIN C, et al. Novel rapid intraoperative qualitative tumor detection by a residual convolutional neural network using label-free stimulated Raman scattering microscopy[J]. Acta Neuropathologica Communications, 2022, 10(1): 109. doi: 10.1186/s40478-022-01411-x
    [33] TASHIBU K. Analysis of water content in rat brain using Raman spectroscopy[J]. No To Shinkei, 1990, 42(10): 999-1004.
    [34] KOLJENOVIĆ S, CHOO-SMITH L P, SCHUT T C B, et al. Discriminating vital tumor from necrotic tissue in human glioblastoma tissue samples by Raman spectroscopy[J]. Laboratory Investigation, 2002, 82(10): 1265-1277. doi: 10.1097/01.LAB.0000032545.96931.B8
    [35] 付荣荣, 鲍甜恬, 田永胜, 等. 基于子成分分解的脑电信号去噪方法比较研究[J]. 计量学报,2019,40(4):708-713. doi: 10.3969/j.issn.1000-1158.2019.04.27

    FU R R, BAO T T, TIAN Y SH, et al. Comparative study on denoising methods of EEG signals based on subcomponent decomposition[J]. Acta Metrologica Sinica, 2019, 40(4): 708-713. (in Chinese). doi: 10.3969/j.issn.1000-1158.2019.04.27
    [36] KAST R E, AUNER G W, ROSENBLUM M L, et al. Raman molecular imaging of brain frozen tissue sections[J]. Journal of Neuro-Oncology, 2014, 120(1): 55-62. doi: 10.1007/s11060-014-1536-9
    [37] LOUIS D N, OHGAKI H, WIESTLER O D, et al. The 2007 WHO classification of tumours of the central nervous system[J]. Acta Neuropathologica, 2007, 114(2): 97-109. doi: 10.1007/s00401-007-0243-4
    [38] BELEITES C, GEIGER K, KIRSCH M, et al. Raman spectroscopic grading of astrocytoma tissues: using soft reference information[J]. Analytical and Bioanalytical Chemistry, 2011, 400(9): 2801-2816. doi: 10.1007/s00216-011-4985-4
    [39] UCKERMANN O, YAO W M, JURATLI T A, et al. IDH1 mutation in human glioma induces chemical alterations that are amenable to optical Raman spectroscopy[J]. Journal of Neuro-Oncology, 2018, 139(2): 261-268.
    [40] RIVA M, SCIORTINO T, SECOLI R, et al. Glioma biopsies classification using Raman spectroscopy and machine learning models on fresh tissue samples[J]. Cancers, 2021, 13(5): 1073. doi: 10.3390/cancers13051073
    [41] UPRETI K, VERMA M, AGRAWAL M, et al. Prediction of mechanical strength by using an artificial neural network and random forest algorithm[J]. Journal of Nanomaterials, 2022, 2022: 7791582.
    [42] KIRSCH M, SCHACKERT G, SALZER R, et al. Raman spectroscopic imaging for in vivo detection of cerebral brain metastases[J]. Analytical and Bioanalytical Chemistry, 2010, 398(4): 1707-1713. doi: 10.1007/s00216-010-4116-7
    [43] BANERJEE H, VERMA M. Intraoperative brain cancer detection with Raman spectroscopy in humans[J]. Annals of Translational Medicine, 2016, 4(4): 68.
    [44] JERMYN M, DESROCHES J, MERCIER J, et al. Raman spectroscopy detects distant invasive brain cancer cells centimeters beyond MRI capability in humans[J]. Biomedical Optics Express, 2016, 7(12): 5129-5137. doi: 10.1364/BOE.7.005129
    [45] DESROCHES J, JERMYN M, PINTO M, et al. A new method using Raman spectroscopy for in vivo targeted brain cancer tissue biopsy[J]. Scientific Reports, 2018, 8(1): 1792. doi: 10.1038/s41598-018-20233-3
    [46] DESROCHES J, LEMOINE É, PINTO M, et al. Development and first in-human use of a Raman spectroscopy guidance system integrated with a brain biopsy needle[J]. Journal of Biophotonics, 2019, 12(3): e201800396. doi: 10.1002/jbio.201800396
    [47] ZHOU Y, LIU CH H, WU B L, et al. Optical biopsy identification and grading of gliomas using label-free visible resonance Raman spectroscopy[J]. Journal of Biomedical Optics, 2019, 24(9): 095001.
    [48] EVANS C L, XU X Y, KESARI S, et al. Chemically-selective imaging of brain structures with CARS microscopy[J]. Optics Express, 2007, 15(19): 12076-12087. doi: 10.1364/OE.15.012076
    [49] CAMP C H JR, LEE Y J, HEDDLESTON J M, et al. High-speed coherent Raman fingerprint imaging of biological tissues[J]. Nature Photonics, 2014, 8(8): 627-634. doi: 10.1038/nphoton.2014.145
    [50] FREUDIGER C W, MIN W, SAAR B G, et al. Label-free biomedical imaging with high sensitivity by stimulated Raman scattering microscopy[J]. Science, 2008, 322(5909): 1857-1861. doi: 10.1126/science.1165758
    [51] JI M B, LEWIS S, CAMELO-PIRAGUA S, et al. Detection of human brain tumor infiltration with quantitative stimulated Raman scattering microscopy[J]. Science Translational Medicine, 2015, 7(309): 309ra163.
    [52] LI J W, WANG CH D, YAO Y, et al. Label-free discrimination of glioma brain tumors in different stages by surface enhanced Raman scattering[J]. Talanta, 2020, 216: 120983. doi: 10.1016/j.talanta.2020.120983
    [53] TANAHASHI K, NATSUME A, OHKA F, et al. Assessment of tumor cells in a mouse model of diffuse infiltrative glioma by Raman spectroscopy[J]. BioMed research international, 2014, 2014: 860241.
    [54] BARKUR S, BANKAPUR A, PRADHAN M, et al. Probing differentiation in cancer cell lines by single-cell micro-Raman spectroscopy[J]. Journal of Biomedical Optics, 2015, 20(8): 085001. doi: 10.1117/1.JBO.20.8.085001
    [55] ITURRIOZ-RODRÍGUEZ N, DE PASQUALE D, FIASCHI P, et al. Discrimination of glioma patient-derived cells from healthy astrocytes by exploiting Raman spectroscopy[J]. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 2022, 269: 120773. doi: 10.1016/j.saa.2021.120773
    [56] ZHAO J, LIU X Y, ZHOU Y, et al. Surface-enhanced Raman scattering technology based on TiO2/Nb2C coated microfluidic chip for monitoring glioma cells invasion in real time[J]. Chinese Chemical Letters, 2023, 34(6): 107895. doi: 10.1016/j.cclet.2022.107895
    [57] LARION M, DOWDY T, RUIZ-RODADO V, et al. Detection of metabolic changes induced via drug treatments in live cancer cells and tissue using Raman imaging microscopy[J]. Biosensors, 2018, 9(1): 5. doi: 10.3390/bios9010005
    [58] YUAN Y H, SHAH N, ALMOHAISIN M I, et al. Assessing fatty acid-induced lipotoxicity and its therapeutic potential in glioblastoma using stimulated Raman microscopy[J]. Scientific Reports, 2021, 11(1): 7422. doi: 10.1038/s41598-021-86789-9
    [59] ZACHARIAH M A, OLIVEIRA-COSTA J P, CARTER B S, et al. Blood-based biomarkers for the diagnosis and monitoring of gliomas[J]. Neuro-Oncology, 2018, 20(9): 1155-1161. doi: 10.1093/neuonc/noy074
    [60] SILANTYEV A S, FALZONE L, LIBRA M, et al. Current and future trends on diagnosis and prognosis of glioblastoma: from molecular biology to proteomics[J]. Cells, 2019, 8(8): 863. doi: 10.3390/cells8080863
    [61] SURMAN M, STĘPIEŃ E, HOJA-ŁUKOWICZ D, et al. Deciphering the role of ectosomes in cancer development and progression: focus on the proteome[J]. Clinical & Experimental Metastasis, 2017, 34(3-4): 273-289.
    [62] GOURLAY J, MOROKOFF A P, LUWOR R B, et al. The emergent role of exosomes in glioma[J]. Journal of Clinical Neuroscience, 2017, 35: 13-23. doi: 10.1016/j.jocn.2016.09.021
    [63] MANGANO K, MAZZON E, BASILE M S, et al. Pathogenic role for macrophage migration inhibitory factor in glioblastoma and its targeting with specific inhibitors as novel tailored therapeutic approach[J]. Oncotarget, 2018, 9(25): 17951-17970. doi: 10.18632/oncotarget.24885
    [64] NAKAGAWA H, YAMADA M, KANAYAMA T, et al. Myelin basic protein in the cerebrospinal fluid of patients with brain tumors[J]. Neurosurgery, 1994, 34(5): 825-833.
    [65] KWON H, OH S, JIN X, et al. Cancer metabolomics in basic science perspective[J]. Archives of Pharmacal Research, 2015, 38(3): 372-380. doi: 10.1007/s12272-015-0552-4
    [66] ALI J S, AIN N U, NAZ S, et al. Biomarker selection and imaging design in cancer: a link with biochemical pathways for imminent engineering[J]. Heliyon, 2020, 6(2): e03340. doi: 10.1016/j.heliyon.2020.e03340
    [67] BULIK M, JANCALEK R, VANICEK J, et al. Potential of MR spectroscopy for assessment of glioma grading[J]. Clinical Neurology and Neurosurgery, 2013, 115(2): 146-153. doi: 10.1016/j.clineuro.2012.11.002
    [68] STADLBAUER A, GRUBER S, NIMSKY C, et al. Preoperative grading of gliomas by using metabolite quantification with high-spatial-resolution proton MR spectroscopic imaging[J]. Radiology, 2006, 238(3): 958-969. doi: 10.1148/radiol.2382041896
    [69] FUJITA Y, KOHTA M, SASAYAMA T, et al. Intraoperative 3-T magnetic Resonance spectroscopy for detection of proliferative remnants of glioma[J]. World Neurosurgery, 2020, 137: 149-157. doi: 10.1016/j.wneu.2020.01.217
    [70] MÖRÉN L, BERGENHEIM A T, GHASIMI S, et al. Metabolomic screening of tumor tissue and serum in glioma patients reveals diagnostic and prognostic information[J]. Metabolites, 2015, 5(3): 502-520. doi: 10.3390/metabo5030502
    [71] KRUCHKO C, GITTLEMAN H, RUHL J, et al. Cancer collection efforts in the United States provide clinically relevant data on all primary brain and other CNS tumors[J]. Neuro-Oncology Practice, 2019, 6(5): 330-339. doi: 10.1093/nop/npz029
    [72] DANG L, WHITE D W, GROSS S, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate[J]. Nature, 2010, 465(7300): 966-966.
    [73] MIAO X X, FANG Q Q, XIAO X, et al. Integrating cycled enzymatic DNA amplification and surface-enhanced Raman scattering for sensitive detection of circulating tumor DNA[J]. Frontiers in Molecular Biosciences, 2021, 8: 676065. doi: 10.3389/fmolb.2021.676065
    [74] ZHOU Y, LIU CH H, WU B L, et al. Invited article: molecular biomarkers characterization for human brain glioma grading using visible resonance Raman spectroscopy[J]. APL Photonics, 2018, 3(12): 120802. doi: 10.1063/1.5036637
    [75] QUESNEL A, COLES N, ANGIONE C, et al. Glycosylation spectral signatures for glioma grade discrimination using Raman spectroscopy[J]. BMC Cancer, 2023, 23(1): 174. doi: 10.1186/s12885-023-10588-w
    [76] GE M L, WANG Y Y, ZHANG F, et al. Study of low-frequency spectroscopic characteristics of γ-aminobutyric acid with THz and low-wavenumber Raman spectroscopy[J]. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 2024, 305: 123550. doi: 10.1016/j.saa.2023.123550
  • 加载中
图(22) / 表(2)
计量
  • 文章访问数:  83
  • HTML全文浏览量:  51
  • PDF下载量:  7
  • 被引次数: 0
出版历程
  • 收稿日期:  2024-01-02
  • 录用日期:  2024-02-26
  • 网络出版日期:  2024-05-10

目录

    /

    返回文章
    返回