Journal Home > Online First

Morphological imaging techniques are very limited in their ability to evaluate the early efficacy of tumor therapies, with the limitation of being more reflective and lagging. Many of the newer therapies are cytostatic, and tumor necrosis or lack of tumor progression is associated with a good response to treatment even in the absence of tumor shrinkage; therefore, there is an increasing need to develop techniques for the assessment of tumor efficacy. Magnetic resonance imaging (MRI), with the help of a variety of contrast mechanisms and probes, provides excellent soft-tissue imaging, high-quality anatomical signals as well as reflecting certain functional states of the tumor and molecular biological information. It can be used to differentiate between cancer and normal tissue, to noninvasively monitor tumor growth, and to identify changes in the tumor and its microenvironment in response to treatment. This review will discuss the role of magnetic resonance imaging in the assessment of tumor efficacy, with a focus on presenting research advances in magnetic resonance molecular imaging and its probes in the assessment of tumor efficacy.


menu
Abstract
Full text
Outline
About this article

Magnetic Resonance Imaging and Its Molecular Probes in Evaluating the Response to Tumor Treatment

Show Author's information Dinghua LiuWeitao YangBingbo Zhang( )
Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai 200065, China

Abstract

Morphological imaging techniques are very limited in their ability to evaluate the early efficacy of tumor therapies, with the limitation of being more reflective and lagging. Many of the newer therapies are cytostatic, and tumor necrosis or lack of tumor progression is associated with a good response to treatment even in the absence of tumor shrinkage; therefore, there is an increasing need to develop techniques for the assessment of tumor efficacy. Magnetic resonance imaging (MRI), with the help of a variety of contrast mechanisms and probes, provides excellent soft-tissue imaging, high-quality anatomical signals as well as reflecting certain functional states of the tumor and molecular biological information. It can be used to differentiate between cancer and normal tissue, to noninvasively monitor tumor growth, and to identify changes in the tumor and its microenvironment in response to treatment. This review will discuss the role of magnetic resonance imaging in the assessment of tumor efficacy, with a focus on presenting research advances in magnetic resonance molecular imaging and its probes in the assessment of tumor efficacy.

Keywords: magnetic resonance imaging (MRI), tumor treatment, molecular probes, efficacy assessment

References(147)

[1]

R.L. Siegel, K.D. Miller, H.E. Fuchs, et al. Cancer statistics, 2022. CA:A Cancer Journal for Clinicians, 2022, 72(1): 7−33. https://doi.org/10.3322/caac.21708

[2]

R.L. Wahl, H. Jacene, Y. Kasamon, et al. From RECIST to PERCIST: Evolving considerations for PET response criteria in solid tumors. Journal of Nuclear Medicine, 2009, 50(Suppl 1): 122S−150S. https://doi.org/10.2967/jnumed.108.057307

[3]

M.K. Palmer. WHO handbook for reporting results of cancer treatment. British Journal of Cancer, 1982, 45(3): 484−485. https://doi.org/10.1038/bjc.1982.83

[4]

P. Therasse, S.G. Arbuck, E.A. Eisenhauer, et al. New guidelines to evaluate the response to treatment in solid tumors. JNCI:Journal of the National Cancer Institute, 2000, 92(3): 205−216. https://doi.org/10.1093/jnci/92.3.205

[5]

E.A. Eisenhauer, P. Therasse, J. Bogaerts, et al. New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). European Journal of Cancer, 2009, 45(2): 228−247. https://doi.org/10.1016/j.ejca.2008.10.026

[6]

C.C. Ko, L.R. Yeh, Y.T. Kuo, et al. Imaging biomarkers for evaluating tumor response: RECIST and beyond. Biomarker Research, 2021, 9(1): 52. https://doi.org/10.1186/s40364-021-00306-8

[7]

S. Saini. Radiologic measurement of tumor size in clinical trials. American Journal of Roentgenology, 2001, 176(2): 333−334. https://doi.org/10.2214/ajr.176.2.1760333

[8]

M.J. Ratain, S.G. Eckhardt. Phase II studies of modern drugs directed against new targets: If you are fazed, too, then resist RECIST. Journal of Clinical Oncology, 2004, 22(22): 4442−4445. https://doi.org/10.1200/jco.2004.07.960

[9]

T.C. Kwee, T. Takahara, D.W.J. Klomp, et al. Cancer imaging: Novel concepts in clinical magnetic resonance imaging. Journal of Internal Medicine, 2010, 268(2): 120−132. https://doi.org/10.1111/j.1365-2796.2010.02243.x

[10]

D.E. Lee, H. Koo, I.C. Sun, et al. Multifunctional nanoparticles for multimodal imaging and theragnosis. Chemical Society Reviews, 2012, 41(7): 2656−2672. https://doi.org/10.1039/c2cs15261d

[11]

S.J. Gwyther, L.H. Schwartz. How to assess anti-tumour efficacy by imaging techniques. European Journal of Cancer, 2008, 44(1): 39−45. https://doi.org/10.1016/j.ejca.2007.10.010

[12]

B.D. Chen, L.H. Liu, R.Y. Yue, et al. Stimuli-responsive switchable MRI nanoprobe for tumor theranostics. Nano Today, 2023, 51: 101931. https://doi.org/10.1016/j.nantod.2023.101931

[13]

E. Terreno, D.D. Castelli, A. Viale, et al. Challenges for molecular magnetic resonance imaging. Chemical Reviews, 2010, 110(5): 3019−3042. https://doi.org/10.1021/cr100025t

[14]
J. Wahsner, , Gale, E. M., Rodríguez-Rodríguez, A., Caravan, P. Chemistry of MRI contrast agents: Current challenges and new frontiers. Chemical Reviews, 2019, 119(2): 957–1057.
DOI
[15]

X. Jin, W.T. Yang, Y. Xu, wt al. Emerging strategies of activatable MR imaging probes and their advantages for biomedical applications. VIEW, 2021, 2(5): 20200141. https://doi.org/10.1002/viw.20200141

[16]

R. Glynne-Jones, L. Wyrwicz, E. Tiret, et al. Rectal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology, 2017, 28: iv22−iv40. https://doi.org/10.1093/annonc/mdx224

[17]

T. Anani, S. Rahmati, N. Sultana, et al. MRI-traceable theranostic nanoparticles for targeted cancer treatment. Theranostics, 2021, 11(2): 579−601. https://doi.org/10.7150/thno.48811

[18]

D. Leung, X. Han, T. Mikkelsen, et al. Role of MRI in primary brain tumor evaluation. Journal of National Comprehensive Cancer Network, 2014, 12(11): 1561−1568. https://doi.org/10.6004/jnccn.2014.0156

[19]

B.B. Kasten, N. Udayakumar, J.W. Leavenworth, et al. Current and future imaging methods for evaluating response to immunotherapy in neuro-oncology. Theranostics, 2019, 9(17): 5085−5104. https://doi.org/10.7150/thno.34415

[20]

D.A. Torigian, S.S. Huang, M. Houseni, et al. Functional imaging of cancer with emphasis on molecular techniques. CA:A Cancer Journal for Clinicians, 2007, 57(4): 206−224. https://doi.org/10.3322/canjclin.57.4.206

[21]
A. Dzik-Jurasz, C. Domenig, M. George, et al. Diffusion MRI for prediction of response of rectal cancer to chemoradiation. The Lancet, 2002, 360(9329): 307–308.
DOI
[22]

L. Kwock, J.K. Smith, M. Castillo, et al. Clinical role of proton magnetic resonance spectroscopy in oncology: Brain, breast, and prostate cancer. The Lancet Oncology, 2006, 7(10): 859−868. https://doi.org/10.1016/s1470-2045(06)70905-6

[23]

A.R. Padhani, C. Hayes, L. Assersohn, et al. Prediction of clinicopathologic response of breast cancer to primary chemotherapy at contrast-enhanced MR imaging: Initial clinical results. Radiology, 2006, 239(2): 361−374. https://doi.org/10.1148/radiol.2392021099

[24]

J.Y. Liang, Q.Q. Cheng, J.X. Huang, et al. Monitoring tumour microenvironment changes during anti-angiogenesis therapy using functional MRI. Angiogenesis, 2019, 22(3): 457−470. https://doi.org/10.1007/s10456-019-09670-4

[25]

F. Giganti, A. Ambrosi, D. Chiari, et al. Apparent diffusion coefficient by diffusion-weighted magnetic resonance imaging as a sole biomarker for staging and prognosis of gastric cancer. Chinese Journal of Cancer Research, 2017, 29(2): 118−126. https://doi.org/10.21147/j.issn.1000-9604.2017.02.04

[26]
T.L. Chenevert, C.R. Meyer, B.A. Moffat, et al. Diffusion MRI: A new strategy for assessment of cancer therapeutic efficacy. Molecular Imaging, 2002, 1(4).
DOI
[27]

R.L. McLaughlin, D.C. Newitt, L.J. Wilmes, et al. High resolution in vivo characterization of apparent diffusion coefficient at the tumor-stromal boundary of breast carcinomas: A pilot study to assess treatment response using proximity-dependent diffusion-weighted imaging. Journal of Magnetic Resonance Imaging, 2014, 39(5): 1308−1313. https://doi.org/10.1002/jmri.24283

[28]

X.D. Shi, W.T. Yang, Q. Ma, et al. Hemoglobin-mediated biomimetic synthesis of paramagnetic O2-evolving theranostic nanoprobes for MR imaging-guided enhanced photodynamic therapy of tumor. Theranostics, 2020, 10(25): 11607−11621. https://doi.org/10.7150/thno.46228

[29]

P. Jagoda, J. Fleckenstein, M. Sonnhoff, et al. Diffusion-weighted MRI improves response assessment after definitive radiotherapy in patients with NSCLC. Cancer Imaging, 2021, 21(1): 15. https://doi.org/10.1186/s40644-021-00384-9

[30]

V. Zampa, G. Aringhieri, R. Tintori, et al. The added value of the visual analysis of DWI in post-surgery follow-up of soft tissue sarcoma of the extremities: Do we really need ADC. La Radiologia Medica, 2023, 128(4): 467−479. https://doi.org/10.1007/s11547-023-01613-w

[31]

P. Ditter, E. Hattingen. MR-spectroscopy in brain tumors. Radiologe, 2017, 57(6): 450−458. https://doi.org/10.1007/s00117-017-0241-z

[32]

J.R. Roebuck, K.M. Cecil, M.D. Schnall, et al. Human breast lesions: Characterization with proton MR spectroscopy. Radiology, 1998, 209(1): 269−275. https://doi.org/10.1148/radiology.209.1.9769842

[33]

K.M. Cecil. Proton magnetic resonance spectroscopy technique for the neuroradiologist. Neuroimaging Clinics of North America, 2013, 23(3): 381−392. https://doi.org/10.1016/j.nic.2012.10.003

[34]
M.F. Kircher, J.R. Allport, E.E. Graves, et al. In vivo high resolution three-dimensional imaging of antigen-specific cytotoxic T-lymphocyte trafficking to tumors. Cancer Research, 2003, 63(20): 6838–6846.
[35]

A. Saha, K.K. Peck, E. Lis, et al. Magnetic resonance perfusion characteristics of hypervascular renal and hypovascular prostate spinal metastases. Spine, 2014, 39(24): E1433−E1440. https://doi.org/10.1097/brs.0000000000000570

[36]

T. Christen, B. Lemasson, N. Pannetier, et al. Is T2* enough to assess oxygenation? quantitative blood oxygen level–dependent analysis in brain tumor. Radiology, 2012, 262(2): 495−502. https://doi.org/10.1148/radiol.11110518

[37]

J.B. Cao, H.Q. An, X.L. Huang, et al. Monitoring of the tumor response to nano-graphene oxide-mediated photothermal/photodynamic therapy by diffusion-weighted and BOLD MRI. Nanoscale, 2016, 8(19): 10152−10159. https://doi.org/10.1039/c6nr02012g

[38]

S. Gross, A. Gilead, A. Scherz, et al. Monitoring photodynamic therapy of solid tumors online by BOLD-contrast MRI. Nature Medicine, 2003, 9(10): 1327−1331. https://doi.org/10.1038/nm940

[39]

A.A. Gilad, T. Israely, H. Dafni. et al. Functional and molecular mapping of uncoupling between vascular permeability and loss of vascular maturation in ovarian carcinoma xenografts: The role of stroma cells in tumor angiogenesis. International Journal of Cancer, 2005, 117(2): 202−211. https://doi.org/10.1002/ijc.21179

[40]

S.P. Rowe, M.G. Pomper. Molecular imaging in oncology: Current impact and future directions. CA:A Cancer Journal for Clinicians, 2022, 72(4): 333−352. https://doi.org/10.3322/caac.21713

[41]

R. Weissleder, M.J. Pittet. Imaging in the era of molecular oncology. Nature, 2008, 452(7187): 580−589. https://doi.org/10.1038/nature06917

[42]

R. Weissleder. Molecular imaging in cancer. Science, 2006, 312(5777): 1168−1171. https://doi.org/10.1126/science.1125949

[43]

W. Reichardt, D. von Elverfeldt. Preclinical applications of magnetic resonance imaging in oncology. Recent Results in Cancer Research Fortschritte Der Krebsforschung Progres Dans Les Recherches Sur Le Cancer, 2020, 216: 405−437. https://doi.org/10.1007/978-3-030-42618-7_12

[44]

Z.R. Lu, V. Laney, Y.J. Li. Targeted contrast agents for magnetic resonance molecular imaging of cancer. Accounts of Chemical Research, 2022, 55(19): 2833−2847. https://doi.org/10.1021/acs.accounts.2c00346

[45]

B.A. Carneiro, W.S. El-Deiry. Targeting apoptosis in cancer therapy. Nature Reviews Clinical Oncology, 2020, 17(7): 395−417. https://doi.org/10.1038/s41571-020-0341-y

[46]

A. Najafov, H.B. Chen, J.Y. Yuan. Necroptosis and cancer. Trends in Cancer, 2017, 3(4): 294−301. https://doi.org/10.1016/j.trecan.2017.03.002

[47]

Y.P. Wang, W.Q. Gao, X.Y. Shi, et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature, 2017, 547(7661): 99−103. https://doi.org/10.1038/nature22393

[48]

J. Debnath, N. Gammoh, K.M. Ryan. Autophagy and autophagy-related pathways in cancer. Nature Reviews Molecular Cell Biology, 2023, 24(8): 560−575. https://doi.org/10.1038/s41580-023-00585-z

[49]

J.P. Friedmann Angeli, D.V. Krysko, M. Conrad. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nature Reviews Cancer, 2019, 19(7): 405−414. https://doi.org/10.1038/s41568-019-0149-1

[50]

P. Tsvetkov, S. Coy, B. Petrova, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science, 2022, 375(6586): 1254−1261. https://doi.org/10.1126/science.abf0529

[51]

G. Pistritto, D. Trisciuoglio, C. Ceci, et al. Apoptosis as anticancer mechanism: Function and dysfunction of its modulators and targeted therapeutic strategies. Aging, 2016, 8(4): 603−619. https://doi.org/10.18632/aging.100934

[52]

S. Fulda, K.M. Debatin. Targeting apoptosis pathways in cancer therapy. Current Cancer Drug Targets, 2004, 4(7): 569−576. https://doi.org/10.2174/1568009043332763

[53]

V.A. Fadok, D.L. Bratton, D.M. Rose, et al. A receptor for phosphatidylserine-specific clearance of apoptotic cells. Nature, 2000, 405(6782): 85−90. https://doi.org/10.1038/35011084

[54]
G. Odabaei, D. Chatterjee, A.R. Jazirehi, et al. Raf-1 kinase inhibitor protein: Structure, function, regulation of cell signaling, and pivotal role in apoptosis. In: Advances in Cancer Research. Amsterdam: Elsevier, 2004: 169–200. https://doi.org/10.1016/s0065-230x(04)91005-6
DOI
[55]

A.A. Rybczynska, H.H. Boersma, S. de Jong, et al. Avenues to molecular imaging of dying cells: Focus on cancer. Medicinal Research Reviews, 2018, 38(6): 1713−1768. https://doi.org/10.1002/med.21495

[56]

D.J. Zhang, Q.M. Jin, C.H. Jiang, et al. Imaging cell death: Focus on early evaluation of tumor response to therapy. Bioconjugate Chemistry, 2020, 31(4): 1025−1051. https://doi.org/10.1021/acs.bioconjchem.0c00119

[57]

H.L. Xu, K.L. Mao, Y.P. Huang, et al. Glioma-targeted superparamagnetic iron oxide nanoparticles as drug-carrying vehicles for theranostic effects. Nanoscale, 2016, 8(29): 14222−14236. https://doi.org/10.1039/c6nr02448c

[58]

S. Shalini, L. Dorstyn, S. Dawar, et al. Old, new and emerging functions of caspases. Cell Death &Differentiation, 2015, 22(4): 526−539. https://doi.org/10.1038/cdd.2014.216

[59]

B. Yoo, M.D. Pagel. A PARACEST MRI contrast agent to detect enzyme activity. Journal of the American Chemical Society, 2006, 128(43): 14032−14033. https://doi.org/10.1021/ja063874f

[60]

D.J. Ye, A.J. Shuhendler, P. Pandit, et al. Caspase-responsive smart gadolinium-based contrast agent for magnetic resonance imaging of drug-induced apoptosis. Chemical Science, 2014, 5(10): 3845−3852. https://doi.org/10.1039/c4sc01392a

[61]

Y. Yuan, Z.L. Ding, J.C. Qian, et al. Casp3/7-instructed intracellular aggregation of Fe3O4 nanoparticles enhances T2 MR imaging of tumor apoptosis. Nano Letters, 2016, 16(4): 2686−2691. https://doi.org/10.1021/acs.nanolett.6b00331

[62]

H. Li, G. Parigi, C. Luchinat, et al. Bimodal fluorescence-magnetic resonance contrast agent for apoptosis imaging. Journal of the American Chemical Society, 2019, 141(15): 6224−6233. https://doi.org/10.1021/jacs.8b13376

[63]

H. Pelicano, D. Carney, P. Huang. ROS stress in cancer cells and therapeutic implications. Drug Resistance Updates, 2004, 7(2): 97−110. https://doi.org/10.1016/j.drup.2004.01.004

[64]

C. Gorrini, I.S. Harris, T.W. Mak. Modulation of oxidative stress as an anticancer strategy. Nature Reviews Drug Discovery, 2013, 12(12): 931−947. https://doi.org/10.1038/nrd4002

[65]

B.L. Dai, R. Zhang, S.H. Qi, et al. Intravital molecular imaging reveals that ROS-caspase-3-GSDME-induced cell punching enhances humoral immunotherapy targeting intracellular tumor antigens. Theranostics, 2022, 12(17): 7603−7623. https://doi.org/10.7150/thno.75966

[66]
G. Bačić, A. Pavićević, F. Peyrot. In vivo evaluation of different alterations of redox status by studying pharmacokinetics of nitroxides using magnetic resonance techniques. Redox Biology, 2016, 8: 226–242. https://doi.org/10.1016/j.redox.2015.10.007
DOI
[67]

H. Wang, D.Q. Yu, B. Li, et al. Ultrasensitive magnetic resonance imaging of systemic reactive oxygen species in vivo for early diagnosis of sepsis using activatable nanoprobes. Chemical Science, 2019, 10(13): 3770−3778. https://doi.org/10.1039/c8sc04961k

[68]
X.L.Li, Y. Liu, X.W. Qi, et al. Sensitive activatable nanoprobes for real-time ratiometric magnetic resonance imaging of reactive oxygen species and ameliorating inflammation in vivo. Advanced Materials, 2022, 34(19): 2109004.
DOI
[69]

Z.J. Zhou, H.Z. Deng, W.J. Yang, et al. Early stratification of radiotherapy response by activatable inflammation magnetic resonance imaging. Nature Communications, 2020, 11: 3032. https://doi.org/10.1038/s41467-020-16771-y

[70]
K. Deng, B. Wu, C.X. Wang, et al. An oxidation-enhanced magnetic resonance imaging probe for visual and specific detection of singlet oxygen generated in photodynamic cancer therapy in vivo. Advanced Healthcare Materials, 2020, 9(16): 2000533.
DOI
[71]

D. Trachootham, J. Alexandre, P. Huang. Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach. Nature Reviews Drug Discovery, 2009, 8(7): 579−591. https://doi.org/10.1038/nrd2803

[72]

S. Dixon, K. Lemberg, M. Lamprecht, et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell, 2012, 149(5): 1060−1072. https://doi.org/10.1016/j.cell.2012.03.042

[73]

S.J. Dixon, B.R. Stockwell. The role of iron and reactive oxygen species in cell death. Nature Chemical Biology, 2014, 10(1): 9−17. https://doi.org/10.1038/nchembio.1416

[74]

X. Chen, R. Kang, G. Kroemer, et al. Broadening horizons: The role of ferroptosis in cancer. Nature Reviews Clinical Oncology, 2021, 18(5): 280−296. https://doi.org/10.1038/s41571-020-00462-0

[75]

G. Lei, L. Zhuang, B.Y. Gan. Targeting ferroptosis as a vulnerability in cancer. Nature Reviews Cancer, 2022, 22(7): 381−396. https://doi.org/10.1038/s41568-022-00459-0

[76]

G.Q. Guan, C. Zhang, H.Y. Liu, et al. Ternary alloy PtWMn as a Mn nanoreservoir for high-field MRI monitoring and highly selective ferroptosis therapy. Angewandte Chemie International Edition, 2022, 61(31): 2117229. https://doi.org/10.1002/anie.202117229

[77]

I. Martínez-Reyes, N.S. Chandel. Cancer metabolism: Looking forward. Nature Reviews Cancer, 2021, 21(10): 669−680. https://doi.org/10.1038/s41568-021-00378-6

[78]

M.G. Vander Heiden, L.C. Cantley, C.B. Thompson. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science, 2009, 324(5930): 1029−1033. https://doi.org/10.1126/science.1160809

[79]

R.A. Cairns, I.S. Harris, T.W. Mak. Regulation of cancer cell metabolism. Nature Reviews Cancer, 2011, 11(2): 85−95. https://doi.org/10.1038/nrc2981

[80]

K. Hönigova, J. Navratil, B. Peltanova, et al. Metabolic tricks of cancer cells. Biochimica et Biophysica - Acta Reviews on Cancer, 2022, 1877(3): 188705. https://doi.org/10.1016/j.bbcan.2022.188705

[81]

O. Jacobson, X.Y. Chen. Interrogating tumor metabolism and tumor microenvironments using molecular positron emission tomography imaging. theranostic approaches to improve therapeutics. Pharmacological Reviews, 2013, 65(4): 1214−1256. https://doi.org/10.1124/pr.113.007625

[82]

M.G. Vander Heiden, R.J. DeBerardinis. Understanding the intersections between metabolism and cancer biology. Cell, 2017, 168(4): 657−669. https://doi.org/10.1016/j.cell.2016.12.039

[83]

A.A. Neves, Y.A. Wainman, A. Wright, et al. Imaging glycosylation in vivo by metabolic labeling and magnetic resonance imaging. Angewandte Chemie International Edition, 2016, 55(4): 1286−1290. https://doi.org/10.1002/anie.201509858

[84]

Y. Yuan, J. Zhang, X.L. Qi, et al. Furin-mediated intracellular self-assembly of olsalazine nanoparticles for enhanced magnetic resonance imaging and tumour therapy. Nature Materials, 2019, 18(12): 1376−1383. https://doi.org/10.1038/s41563-019-0503-4

[85]
Y. Kondo, H. Nonaka, Y. Takakusagi, et al. Design of nuclear magnetic resonance molecular probes for hyperpolarized bioimaging. Angewandte Chemie International Edition, 2021, 60(27): 14779–14799.
DOI
[86]

S. Siddiqui, S. Kadlecek, M. Pourfathi, et al. The use of hyperpolarized carbon-13 magnetic resonance for molecular imaging. Advanced Drug Delivery Reviews, 2017, 113: 3−23. https://doi.org/10.1016/j.addr.2016.08.011

[87]

S.E. Day, M.I. Kettunen, F.A. Gallagher, et al. Detecting tumor response to treatment using hyperpolarized 13C magnetic resonance imaging and spectroscopy. Nature Medicine, 2007, 13(11): 1382−1387. https://doi.org/10.1038/nm1650

[88]

N. Koyasu, F. Hyodo, R. Iwasaki, et al. Spatiotemporal imaging of redox status using in vivo dynamic nuclear polarization magnetic resonance imaging system for early monitoring of response to radiation treatment of tumor. Free Radical Biology and Medicine, 2022, 179: 170−180. https://doi.org/10.1016/j.freeradbiomed.2021.12.311

[89]

P. Carmeliet, R.K. Jain. Angiogenesis in cancer and other diseases. Nature, 2000, 407(6801): 249−257. https://doi.org/10.1038/35025220

[90]

R.A. Sharma, A.L. Harris, A.G. Dalgleish, et al. Angiogenesis as a biomarker and target in cancer chemoprevention. The Lancet Oncology, 2001, 2(12): 726−732. https://doi.org/10.1016/s1470-2045(01)00586-1

[91]

P. Carmeliet, R.K. Jain. Molecular mechanisms and clinical applications of angiogenesis. Nature, 2011, 473(7347): 298−307. https://doi.org/10.1038/nature10144

[92]

A.R. Hsu, X.Y. Chen. Advances in anatomic, functional, and molecular imaging of angiogenesis. Journal of Nuclear Medicine, 2008, 49(4): 511−514. https://doi.org/10.2967/jnumed.107.050179

[93]

Z.Y. Jia, L.N. Song, F.C. Zang, et al. Active-target T1-weighted MR imaging of tiny hepatic tumor via RGD modified ultra-small Fe3O4 nanoprobes. Theranostics, 2016, 6(11): 1780−1791. https://doi.org/10.7150/thno.14280

[94]

A.K. Olsson, A. Dimberg, J. Kreuger, et al. VEGF receptor signalling? in control of vascular function. Nature Reviews Molecular Cell Biology, 2006, 7(5): 359−371. https://doi.org/10.1038/nrm1911

[95]

M.V. Backer, Z. Levashova, V. Patel, et al. Molecular imaging of VEGF receptors in angiogenic vasculature with single-chain VEGF-based probes. Nature Medicine, 2007, 13(4): 504−509. https://doi.org/10.1038/nm1522

[96]

F., Zhang, X.L. Huang, L. Zhu, et al. Noninvasive monitoring of orthotopic glioblastoma therapy response using RGD-conjugated iron oxide nanoparticles. Biomaterials, 2012, 33(21): 5414−5422. https://doi.org/10.1016/j.biomaterials.2012.04.032

[97]

D.B. Ellegala, H. Leong-Poi, J.E. Carpenter, et al. Imaging tumor angiogenesis with contrast ultrasound and microbubbles targeted to alpha(v)beta3. Circulation, 2003, 108(3): 336−341. https://doi.org/10.1161/01.CIR.0000080326.15367.0C

[98]

D.A. Sipkins, D.A. Cheresh, M.R. Kazemi, et al. Detection of tumor angiogenesis in vivo by alphaVbeta3-targeted magnetic resonance imaging. Nature Medicine, 1998, 4(5): 623−626. https://doi.org/10.1038/nm0598-623

[99]

G. Suna, W. Wojakowski, M. Lynch, et al. Extracellular matrix proteomics reveals interplay of aggrecan and aggrecanases in vascular remodeling of stented coronary arteries. Circulation, 2018, 137(2): 166−183. https://doi.org/10.1161/circulationaha.116.023381

[100]

J.O. Kaufmann, J. Brangsch, A. Kader, et al. ADAMTS4-specific MR probe to assess aortic aneurysms in vivo using synthetic peptide libraries. Nature Communications, 2022, 13: 2867. https://doi.org/10.1038/s41467-022-30464-8

[101]
A.Y. Louie, M.M. Hüber, E.T. Ahrens, et al. In vivo visualization of gene expression using magnetic resonance imaging. Nature Biotechnology, 2000, 18(3): 321–325.
DOI
[102]

J.H. Kang, J.K. Chung. Molecular-genetic imaging based on reporter gene expression. Journal of Nuclear Medicine, 2008, 49(Suppl 2): 164S−179S. https://doi.org/10.2967/jnumed.107.045955

[103]

A.A. Gilad, K. Ziv, M.T. McMahon, et al. MRI reporter genes. Jounral of Nuclear Medicine, 2008, 49(12): 1905−1908. https://doi.org/10.2967/jnumed.108.053520

[104]
Arena, F., Singh, J. B., Gianolio, E., Stefanìa, R., Aime, S. β-gal gene expression MRI reporter in melanoma tumor cells. design, synthesis, and in vitro and in vivo testing of a Gd(III) containing probe forming a high relaxivity, melanin-like structure upon β-gal enzymatic activation. Bioconjugate Chemistry, 2011, 22(12): 2625–2635.
DOI
[105]
M.H. Cao, J.J. Mao, X.H. Duan, et al. In vivo tracking of the tropism of mesenchymal stem cells to malignant gliomas using reporter gene-based MR imaging. International Journal of Cancer, 2018, 142(5): 1033–1046.
DOI
[106]

L.B. Kennedy, A.K.S. Salama. A review of cancer immunotherapy toxicity. CA:A Cancer Journal for Clinicians, 2020, 70(2): 86−104. https://doi.org/10.3322/caac.21596

[107]
W.J. Lesterhuis, A. Bosco, M.J. Millward, et al. Dynamic versus static biomarkers in cancer immune checkpoint blockade: Unravelling complexity. Nature Reviews Drug Discovery, 2017, 16(4): 264–272.
DOI
[108]

Z.B. Xiao, E. Puré. Imaging of T-cell responses in the context of cancer immunotherapy. Cancer Immunology Research, 2021, 9(5): 490−502. https://doi.org/10.1158/2326-6066.cir-20-0678

[109]

A.J. Grippin, B. Wummer, T. Wildes, et al. Dendritic cell-activating magnetic nanoparticles enable early prediction of antitumor response with magnetic resonance imaging. ACS Nano, 2019, 13(12): 13884−13898. https://doi.org/10.1021/acsnano.9b05037

[110]

I. Melero, E. Castanon, M. Alvarez, et al. Intratumoural administration and tumour tissue targeting of cancer immunotherapies. Nature Reviews Clinical Oncology, 2021, 18(9): 558−576. https://doi.org/10.1038/s41571-021-00507-y

[111]

J. Levi, T. Lam, S.R. Goth, et al. Imaging of activated T cells as an early predictor of immune response to anti-PD-1 therapy. Cancer Research, 2019, 79(13): 3455−3465. https://doi.org/10.1158/0008-5472.can-19-0267

[112]

C.R. Shi, Q.Y. Zhang, Y.Y. Yao, et al. Targeting the activity of T cells by membrane surface redox regulation for cancer theranostics. Nature Nanotechnology, 2023, 18(1): 86−97. https://doi.org/10.1038/s41565-022-01261-7

[113]

M. Hockel, P. Vaupel. Tumor hypoxia: Definitions and current clinical, biologic, and molecular aspects. JNCI Journal of the National Cancer Institute, 2001, 93(4): 266−276. https://doi.org/10.1093/jnci/93.4.266

[114]

M. Pastorek, V. Simko, M. Takacova, et al. Sulforaphane reduces molecular response to hypoxia in ovarian tumor cells independently of their resistance to chemotherapy. International Journal of Oncology, 2015, 47(1): 51−60. https://doi.org/10.3892/ijo.2015.2987

[115]

P. Vaupel, A. Mayer. Hypoxia in cancer: Significance and impact on clinical outcome. Cancer and Metastasis Reviews, 2007, 26(2): 225−239. https://doi.org/10.1007/s10555-007-9055-1

[116]

W.R. Wilson, M.P. Hay. Targeting hypoxia in cancer therapy. Nature Reviews Cancer, 2011, 11(6): 393−410. https://doi.org/10.1038/nrc3064

[117]

S.K. Chitneni, G.M. Palmer, M.R. Zalutsky, et al. Molecular imaging of hypoxia. Journal of Nuclear Medicine, 2011, 52(2): 165−168. https://doi.org/10.2967/jnumed.110.075663

[118]

J.N. Liu, W.B. Bu, J.L. Shi. Chemical design and synthesis of functionalized probes for imaging and treating tumor hypoxia. Chemical Reviews, 2017, 117(9): 6160−6224. https://doi.org/10.1021/acs.chemrev.6b00525

[119]

J. Liu, H. Cabral, B. Song, et al. Nanoprobe-based magnetic resonance imaging of hypoxia predicts responses to radiotherapy, immunotherapy, and sensitizing treatments in pancreatic tumors. ACS Nano, 2021, 15(8): 13526−13538. https://doi.org/10.1021/acsnano.1c04263

[120]

V. Estrella, T.A. Chen, M. Lloyd, et al. Acidity generated by the tumor microenvironment drives local invasion. Cancer Research, 2013, 73(5): 1524−1535. https://doi.org/10.1158/0008-5472.can-12-2796

[121]

B.A. Webb, M. Chimenti, M.P. Jacobson, et al. Dysregulated pH: A perfect storm for cancer progression. Nature Reviews Cancer, 2011, 11(9): 671−677. https://doi.org/10.1038/nrc3110

[122]

X.M. Zhang, Y.X. Lin, R.J. Gillies. Tumor pH and its measurement. Journal of Nuclear Medicine, 2010, 51(8): 1167−1170. https://doi.org/10.2967/jnumed.109.068981

[123]

P. Mi, D. Kokuryo, H. Cabral, et al. A pH-activatable nanoparticle with signal-amplification capabilities for non-invasive imaging of tumour malignancy. Nature Nanotechnology, 2016, 11(8): 724−730. https://doi.org/10.1038/nnano.2016.72

[124]

X.L. Zhu, X.X. Tang, H.Y. Lin, et al. A fluorinated ionic liquid-based activatable 19F MRI platform detects biological targets. Chem, 2020, 6(5): 1134−1148. https://doi.org/10.1016/j.chempr.2020.01.023

[125]

H. Sato, T. Takino, Y. Okada, et al. A matrix metalloproteinase expressed on the surface of invasive tumour cells. Nature, 1994, 370(6484): 61−65. https://doi.org/10.1038/370061a0

[126]
J. Chang, W.Q. Cai, C.J. Liang, et al. Enzyme-instructed activation of pro-protein therapeutics in vivo. Journal of the American Chemical Society, 2019, 141(45): 18136–18141.
DOI
[127]

M. Egeblad, Z. Werb. New functions for the matrix metalloproteinases in cancer progression. Nature Reviews Cancer, 2002, 2(3): 161−174. https://doi.org/10.1038/nrc745

[128]

J.S. Rao. Molecular mechanisms of glioma invasiveness: The role of proteases. Nature Reviews Cancer, 2003, 3(7): 489−501. https://doi.org/10.1038/nrc1121

[129]

K. Kessenbrock, V. Plaks, Z. Werb. Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell, 2010, 141(1): 52−67. https://doi.org/10.1016/j.cell.2010.03.015

[130]

R. Roy, J. Yang, M.A. Moses. Matrix metalloproteinases As novel biomarker s and potential therapeutic targets in human cancer. Journal of Clinical Oncology, 2009, 27(31): 5287−5297. https://doi.org/10.1200/jco.2009.23.5556

[131]
V. Amirbekian, J.G.S. Aguinaldo, S. Amirbekian, et al. Atherosclerosis and matrix metalloproteinases: Experimental molecular MR imaging in vivo. Radiology, 2009, 251(2): 429–438.
DOI
[132]

E.S. Olson, T. Jiang, T.A. Aguilera, et al. Activatable cell penetrating peptides linked to nanoparticles as dual probes for in vivo fluorescence and MR imaging of proteases. Proceedings of the National Academy of Sciences of the United States of America, 2010, 107(9): 4311−4316. https://doi.org/10.1073/pnas.0910283107

[133]

H. Shi, Y. Sun, R. Yan, et al. Magnetic semiconductor Gd-doping CuS nanoparticles as activatable nanoprobes for bimodal imaging and targeted photothermal therapy of gastric tumors. Nano Letters, 2019, 19(2): 937−947. https://doi.org/10.1021/acs.nanolett.8b04179

[134]

L. Guzman-Rojas, R. Rangel, A. Salameh, et al. Cooperative effects of aminopeptidase N (CD13) expressed by nonmalignant and cancer cells within the tumor microenvironment. Proceedings of the National Academy of Sciences of the United States of America, 2012, 109(5): 1637−1642. https://doi.org/10.1073/pnas.1120790109

[135]

R. Hata, H. Nonaka, Y. Takakusagi, et al. Design of a hyperpolarized molecular probe for detection of aminopeptidase N activity. Angewandte Chemie International Edition, 2016, 55(5): 1765−1768. https://doi.org/10.1002/anie.201509457

[136]
Y. Saito, H. Yatabe, I. Tamura, et al. Structure-guided design enables development of a hyperpolarized molecular probe for the detection of aminopeptidase N activity in vivo. Science Advances, 2022, 8(13): eabj2667.
DOI
[137]

M.F. Kircher, J.K. Willmann. Molecular body imaging: MR imaging, CT, and US. part I. principles. Radiology, 2012, 263(3): 633−643. https://doi.org/10.1148/radiol.12102394

[138]

J.M. Winfield, G.S. Payne, N.M. DeSouza. Functional MRI and CT biomarkers in oncology. European Journal of Nuclear Medicine and Molecular Imaging, 2015, 42(4): 562−578. https://doi.org/10.1007/s00259-014-2979-0

[139]

J.J. Platt, M.L. Ramanathan, R.A. Crosbie, et al. C-reactive protein as a predictor of postoperative infective complications after curative resection in patients with colorectal cancer. Annals of Surgical Oncology, 2012, 19(13): 4168−4177. https://doi.org/10.1245/s10434-012-2498-9

[140]

C.N. Loynachan, A.P. Soleimany, J.S. Dudani, et al. Renal clearable catalytic gold nanoclusters for in vivo disease monitoring. Nature Nanotechnology, 2019, 14(9): 883−890. https://doi.org/10.1038/s41565-019-0527-6

[141]

S.Q. He, J. Song, J.L. Qu, et al. Crucial breakthrough of second near-infrared biological window fluorophores: Design and synthesis toward multimodal imaging and theranostics. Chemical Society Reviews, 2018, 47(12): 4258−4278. https://doi.org/10.1039/c8cs00234g

[142]

J.S. Huang, L.C. Su, C. Xu, et al. Molecular radio afterglow probes for cancer radiodynamic theranostics. Nature Materials, 2023, 22(11): 1421−1429. https://doi.org/10.1038/s41563-023-01659-1

[143]

Y. X. Hu, J.Y. Zhang, Y.X. Miao, et al. Enzyme-mediated in situ self-assembly promotes in vivo bioorthogonal reaction for pretargeted multimodality imaging. Angewandte Chemie International Edition, 2021, 60(33): 18082−18093. https://doi.org/10.1002/anie.202103307

[144]

F. Seith, A. Forschner, B. Weide, et al. Is there a link between very early changes of primary and secondary lymphoid organs in 18F-FDG-PET/MRI and treatment response to checkpoint inhibitor therapy. Journal for ImmunoTherapy of Cancer, 2020, 8(2): e000656. https://doi.org/10.1136/jitc-2020-000656corr1

[145]

C. Bai, Z.Y. Jia, L.N. Song, et al. Time-dependent T1–T2 switchable magnetic resonance imaging realized by c(RGDyK) modified ultrasmall Fe3O4 nanoprobes. Advanced Functional Materials, 2018, 28(32): 1802281. https://doi.org/10.1002/adfm.201802281

[146]

Z.Y. Shen, T.X. Chen, X.H. Ma, et al. Multifunctional theranostic nanoparticles based on exceedingly small magnetic iron oxide nanoparticles for T1-weighted magnetic resonance imaging and chemotherapy. ACS Nano, 2017, 11(11): 10992−11004. https://doi.org/10.1021/acsnano.7b04924

[147]

J. Wang, Y.H. Jia, Q.Y. Wang, et al. An ultrahigh-field-tailored T1T2 dual-mode MRI contrast agent for high-performance vascular imaging. Advanced Materials, 2021, 33(2): 2004917. https://doi.org/10.1002/adma.202004917

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 10 December 2023
Revised: 01 February 2024
Accepted: 27 February 2024
Published: 02 April 2024

Copyright

© The Author(s) 2024.

Acknowledgements

Acknowledgements

This work was supported by the National Natural Science Foundation of China (Grants 82272137, 82272055), National Key Research and Development Project (Grant 2022YFB3804500), Shanghai Municipal Commission of Health and Family Planning Project (Grant 20204Y0032), Shanghai Sailing Program (Grant 21YF1436600), and the Fundamental Research Funds for the Central Universities (Grant 22120220585).

Rights and permissions

This is an open-access article distributed under  the  terms  of  the  Creative  Commons  Attribution  4.0 International  License (CC BY) (http://creativecommons.org/licenses/by/4.0/), which  permits  unrestricted  use,  distribution,  and reproduction in any medium, provided the original author and source are credited.

Return