Monitoring of Current Cancer Therapy by Positron Emission Tomography and Possible Role of Radiomics Assessment
Abstract
:1. Introduction
2. Glucose Metabolism in Cancer Cells
3. Therapy Monitoring with Imaging Biomarkers of FDG-PET
4. Relationship between PD-L1 Expression and Monitoring of Immunotherapy by FDG-PET
5. Limitations and Prospects of Response Evaluation of ICI Therapy by PET
6. Cancer Metabolomics as Target of Therapy and Response Evaluation by PET
7. Possible Role of PET with Radiomics and Artificial Intelligence for Response Evaluation
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
References
- Ehlerding, E.B.; England, C.G.; McNeel, D.G.; Cai, W. Molecular imaging of immunotherapy targets in cancer. J. Nucl. Med. 2016, 57, 1487–1492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schniering, J.; Benešová, M.; Brunner, M.; Stephanie, H.S.; Cohrs, S.; Frauenfelder, T.; Vrugt, B.; Feghali-Bostwick, C.; Schibli, R.; Distler, O.; et al. 18F-AzaFol for detection of folate receptor-β positive macrophages in experimental interstitial lung disease–A proof-of-concept study. Front. Immunol. 2019, 10, 2724. [Google Scholar] [CrossRef] [Green Version]
- Seo, J.W.; Tavaré, R.; Mahakian, L.M.; Matthew, T.; Silvestrini, M.T.; Tam, S.; Ingham, E.S.; Salazar, F.B.; Borowsky, A.D.; Wu, A.M.; et al. CD8+ T-cell density imaging with 64Cu-labeled cys-diabody informs immunotherapy protocols. Clin. Cancer Res. 2018, 24, 4976–4987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martinez, O.; Sosabowski, J.; Maher, J.; Papa, S. New developments in imaging cell-based therapy. J. Nucl. Med. 2019, 60, 730–735. [Google Scholar] [CrossRef] [PubMed]
- Kitagawa, Y.; Sano, K.; Nishizawa, S.; Nakamura, M.; Ogasawara, T.; Sadato, N.; Yonekura, Y. FDG-PET for prediction of tumour aggressiveness and response to intra-arterial chemotherapy and radiotherapy in head and neck cancer. Eur. J. Nucl. Med. Mol. Imaging 2003, 30, 63–71. [Google Scholar] [CrossRef]
- Warburg, O. The metabolism of carcinoma cells. J. Cancer Res. 1925, 9, 148–163. [Google Scholar] [CrossRef] [Green Version]
- Liberti, M.V.; Locasale, J.W. The Warburg effect: How does it benefit cancer cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef] [Green Version]
- Kernstine, K.H.; Faubert, B.; Do, Q.N.; Rogers, T.J.; Hensley, C.T.; Cai, L.; Torrealba, J.; Oliver, D.; Wachsmann, J.W.; Lenkinski, R.E.; et al. Does tumor FDG-PET avidity represent enhanced glycolytic metabolism in non-small cell lung cancer? Ann. Thorac. Surg. 2020, 109, 1019–1025. [Google Scholar] [CrossRef]
- Momcilovic, M.; Jones, A.; Bailey, S.T.; Waldmann, C.M.; .Li, R.; .Lee, J.T.; Abdelhady, G.; Gomez, A.; Holloway, T.; Schmid, E.; et al. In vivo imaging of mitochondrial membrane potential in non-small-cell lung cancer. Nature 2019, 575, 380–384. [Google Scholar] [CrossRef]
- DeBerardinis, R.J.; Navdeep, S.; Chandel, N.S. We need to talk about the Warburg effect. Nat. Metab. 2020, 2, 127–129. [Google Scholar] [CrossRef]
- Hundshammer, C.; Braeuer, M.; Müller, C.A.; Hansen, A.E.; Schillmaier, M.S.; Düwel, S.; Feuerecker, B.; Glaser, S.J.; Haase, A.; Weichert, W.; et al. Simultaneous characterization of tumor cellularity and the Warburg effect with PET, MRI and hyperpolarized 13C-MRSI. Theranostics 2018, 8, 4765–4780. [Google Scholar] [CrossRef] [PubMed]
- Ji, Z.; Long, H.; Hu, Y.; Ji, Z.; Long, H.; Hu, Y.; Qiu, X.; Chen, X.; Li, Z.; Fan, D.; et al. Expression of MDR1, HIF-1α and MRP1 in sacral chordoma and chordoma cell line CM-319. J. Exp. Clin. Cancer Res. 2010, 29, 158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [Green Version]
- Vaupel, P.; Kallinowski, F.; Okunieff, P. Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: A review. Cancer Res. 1989, 49, 6449–6465. [Google Scholar]
- Brown, J.M.; Wilson, W.R. Exploiting tumour hypoxia in cancer treatment. Nat. Rev. Cancer 2004, 4, 437–447. [Google Scholar] [CrossRef]
- Cairns, R.A.; Mak, T.W. The current state of cancer metabolism. Nat. Rev. Cancer 2016, 16, 613–614. [Google Scholar] [CrossRef]
- LaGory, E.L.; Giaccia, A.J. The ever-expanding role of HIF in tumour and stromal biology. Nat. Cell Biol. 2016, 18, 356–365. [Google Scholar] [CrossRef] [Green Version]
- Chaffer, C.L.; San Juan, B.P.; Lim, E.; Weinberg, R.A. EMT, cell plasticity and metastasis. Cancer Metastasis Rev. 2016, 35, 645–654. [Google Scholar] [CrossRef]
- Pastushenko, I.; Brisebarre, A.; Sifrim, A.; Fioramonti, M.; Revenco, T.; Boumahdi, S.; Van Keymeulen, A.; Brown, D.; Moers, V.; Lemaire, S.; et al. Identification of the tumour transition states occurring during EMT. Nature 2018, 556, 463–468. [Google Scholar] [CrossRef]
- Kaira, K.; Endo, M.; Abe, M.; Nakagawa, K.; Ohde, Y.; Okumura, T.; Takahashi, T.; Murakami, H.; Tsuya, A.; Nakamura, Y.; et al. Biologic correlation of 2-[18F]-fluoro-2-deoxy-D-glucose uptake on positron emission tomography in thymic epithelial tumors. J. Clin. Oncol. 2010, 28, 3746–3753. [Google Scholar] [CrossRef]
- Nagao, A.; Kobayashi, M.; Koyasu, S.; Chow, C.C.T.; Harada, H. HIF-1-dependent reprogramming of glucose metabolic pathway of cancer cells and its therapeutic significance. Int. J. Mol. Sci. 2019, 20, 238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oriuchi, N.; Sugawara, S.; Shiga, T. Positron emission tomography for response evaluation in microenvironment-targeted anti-cancer therapy. Biomedicines 2020, 8, 371. [Google Scholar] [CrossRef] [PubMed]
- Lodge, M.A.; Chaudhry, M.A.; Wahl, R.L. Noise considerations for PET quantification using maximum and peak standardized uptake value. J. Nucl. Med. 2012, 53, 1041–1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boellaard, R.; Delgado-Bolton, R.; Oyen, W.J.G.; Giammarile, F.; Tatsch, K.; Eschner, W.; Verzijlbergen, F.J.; Barrington, S.F.; Pike, L.C.; Weber, W.A.; et al. FDG PET/CT: EANM procedure guidelines for tumour imaging: Version 2.0. Eur. J. Nucl. Med. Mol. Imaging 2015, 42, 328–354. [Google Scholar] [CrossRef] [PubMed]
- Hofheinz, F.; Apostolova, I.; Oehme, L.; Kotzerke, J.; van den Hoff, J. Test-retest variability in lesion SUV and lesion SUR in 18F-FDG PET: An analysis of data from two prospective multicenter trials. J. Nucl. Med. 2017, 58, 1770–1775. [Google Scholar] [CrossRef] [Green Version]
- Chen, K.Y.; Cypess, A.M.; Laughlin, M.R.; Haft, C.R.; Hu, H.H.; Bredella, M.A.; Enerbäck, S.; Kinahan, P.E.; Lichtenbelt, W.; Lin, F.I.; et al. Brown adipose reporting criteria in imaging studies (BARCIST 1.0): Recommendations for standardized FDG-PET/CT experiments in humans. Cell Metab. 2016, 24, 210–222. [Google Scholar] [CrossRef] [Green Version]
- Burger, I.A.; Casanova, R.; Steiger, S.; Husmann, L.; Stolzmann, P.; Huellner, M.W.; Curioni, A.; Hillinger, S.; Schmidtlein, C.R.; Soltermann, A. 18F-FDG PET/CT of non-small cell lung carcinoma under neoadjuvant chemotherapy: Background-based adaptive-volume metrics outperform TLG and MTV in predicting histopathologic response. J. Nucl. Med. 2016, 57, 849–854. [Google Scholar] [CrossRef] [Green Version]
- Minamimoto, R.; Takeda, Y.; Hotta, M.; Toyohara, J.; Nakajima, K.; Naka, G.; Sugiyama, H. 18F-FDG and 11C-4DST PET/CT for evaluating response to platinum-based doublet chemotherapy in advanced non-small cell lung cancer: A prospective study. EJNMMI Res. 2019, 9, 4. [Google Scholar] [CrossRef]
- Im, H.J.; Pak, K.; Cheon, G.J.; Kang, K.W.; Kim, S.J.; Kim, I.J.; Chung, J.K.; Kim, E.E.; Lee, D.S. Prognostic value of volumetric parameters of 18F-FDG PET in non-small-cell lung cancer: A meta-analysis. Eur. J. Nucl. Med. Mol. Imaging 2015, 42, 241–251. [Google Scholar] [CrossRef]
- Eisenhauer, E.A.; Therasse, P.; Bogaerts, J.; Schwartz, L.H.; Sargent, D.; Ford, R.; Dancey, J.; Arbuck, S.; Gwyther, S.; Mooney, M.; et al. New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur. J. Cancer 2009, 45, 228–247. [Google Scholar] [CrossRef]
- Young, H.; Baum, R.; Cremerius, U.; Herholz, K.; Hoekstra, O.; Lammertsma, A.A.; Pruim, J.; Price, P. Measurement of clinical and subclinical tumour response using [18F]-fluorodeoxyglucose and positron emission tomography: Review and 1999 EORTC recommendations. European Organization for Research and Treatment of Cancer (EORTC) PET Study Group. Eur. J. Cancer 1999, 35, 1773–1782. [Google Scholar] [CrossRef]
- Wahl, R.L.; Jacene, H.; Kasamon, Y.; Lodge, M.A. From RECIST to PERCIST: Evolving considerations for PET response criteria in solid tumors. J. Nucl. Med. 2009, 50 (Suppl. 1), 122–150. [Google Scholar] [CrossRef] [Green Version]
- Nishino, M.; Giobbie-Hurder, A.; Gargano, M.; Suda, M.; Ramaiya, N.H.; Hodi, F.S. Developing a common language for tumor response to immunotherapy: Immune-related response criteria using unidimensional measurements. Clin. Cancer Res. 2013, 19, 3936–3943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seymour, L.; Bogaerts, J.; Perrone, A.; Ford, R.; Schwartz, L.H.; Mandrekar, S.; Lin, N.U.; Litière, S.; Dancey, J.; Chen, A.; et al. iRECIST: Guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017, 18, e143–e152. [Google Scholar] [CrossRef] [Green Version]
- Ito, K.; Teng, R.; Schöder, H.; Humm, J.L.; Ni, A.; Michaud, L.; Nakajima, R.; Yamashita, R.; Wolchok, J.D.; Weber, W.A. 18F-FDG PET/CT for monitoring of Ipilimumab therapy in patients with metastatic melanoma. J. Nucl. Med. 2019, 60, 335–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sunaga, N.; Oriuchi, N.; Kaira, K.; Yanagitani, N.; Tomizawa, Y.; Hisada, T.; Ishizuka, T.; Endo, K.; Mori, M. Usefulness of FDG-PET for early prediction of the response to gefitinib in non-small cell lung cancer. Lung Cancer 2008, 59, 203–210. [Google Scholar] [CrossRef]
- Desar, I.M.; van Herpen, C.M.; van Laarhoven, H.W.; Barentsz, J.O.; van der Graaf, W.T. Beyond RECIST: Molecular and functional imaging techniques for evaluation of response to targeted therapy. Cancer Treat. Rev. 2009, 35, 309–321. [Google Scholar] [CrossRef]
- Kaira, K.; Higuchi, T.; Naruse, I.; Arisaka, Y.; Tokue, A.; Altan, B.; Suda, S.; Mogi, A.; Shimizu, K.; Sunaga, N.; et al. Metabolic activity by 18F-FDG-PET/CT is predictive of early response after nivolumab in previously treated NSCLC. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 56–66. [Google Scholar] [CrossRef]
- Takada, K.; Toyokawa, G.; Okamoto, T.; Baba, S.; Kozuma, Y.; Matsubara, T.; Haratake, N.; Akamine, T.; Takamori, S.; Katsura, M.; et al. Metabolic characteristics of programmed cell death-ligand 1-expressing lung cancer on 18F-fluorodeoxyglucose positron emission tomography/computed tomography. Cancer Med. 2017, 6, 2552–2561. [Google Scholar] [CrossRef]
- Kasahara, N.; Kaira, K.; Bao, P.; Higuchi, T.; Arisaka, Y.; Erkhem-Ochira, B.; Sunaga, N.; Ohtaki, Y.; Yajima, T.; Kosaka, T.; et al. Correlation of tumor-related immunity with 18F-FDG-PET in pulmonary squamous-cell carcinoma. Lung Cancer 2018, 119, 71–77. [Google Scholar] [CrossRef]
- Kaira, K.; Shimizu, K.; Kitahara, S.; Yajima, T.; Atsumi, J.; Kosaka, T.; Ohtaki, Y.; Higuchi, T.; Oyama, T.; Asao, T.; et al. 2-Deoxy-2-[fluorine-18] fluoro-d-glucose uptake on positron emission tomography is associated with programmed death ligand-1 expression in patients with pulmonary adenocarcinoma. Eur. J. Cancer 2018, 101, 181–190. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; Wang, D.; Sun, Q.; Pu, H.; Wang, Y.; Zhao, S.; Wang, Y.; Zhang, Q. Prognostic significance of PD-L1 expression and 18F-FDG PET/CT in surgical pulmonary squamous cell carcinoma. Oncotarget 2017, 8, 51630–51640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, R.; Zhou, X.; Liu, J.; Huang, G. Relationship between the expression of PD-1/PD-L1 and 18F-FDG uptake in bladder cancer. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 848–854. [Google Scholar] [CrossRef] [PubMed]
- Kasahara, N.; Kaira, K.; Yamaguchi, K.; Masubuchi, H.; Tsurumaki, H.; Hara, K.; Koga, Y.; Sakurai, R.; Higuchi, T.; Handa, T.; et al. Fluorodeoxyglucose uptake is associated with low tumor-infiltrating lymphocyte levels in patients with small cell lung cancer. Lung Cancer 2019, 134, 180–186. [Google Scholar] [CrossRef]
- Hu, B.; Chen, W.; Zhang, Y.; Shi, H.; Cheng, D.; Xiu, Y. 18F-FDG maximum standard uptake value predicts PD-L1 expression on tumor cells or tumor-infiltrating immune cells in non-small cell lung cancer. Ann. Nucl. Med. 2020, 34, 322–328. [Google Scholar] [CrossRef]
- Jiang, H.; Zhang, R.; Jiang, H.; Zhang, M.; Guo, W.; Zhang, J.; Zhou, X.; Pan, W.; Zhao, S.; Li, P. Retrospective analysis of the prognostic value of PD-L1 expression and 18F-FDG PET/CT metabolic parameters in colorectal cancer. J. Cancer 2020, 11, 2864–2873. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Zhao, N.; Wu, Z.; Pan, N.; Shen, X.; Liu, T.; Wei, F.; You, J.; Xu, W.; Ren, X. New insight on the correlation of metabolic status on 18F-FDG PET/CT with immune marker expression in patients with non-small cell lung cancer. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 1127–1136. [Google Scholar] [CrossRef]
- Zhao, L.; Zhuang, Y.; Fu, K.; Chen, P.; Wang, Y.; Zhuo, J.; Liao, X.; Chen, H.; Lin, Q. Usefulness of [18F]fluorodeoxyglucose PET/CT for evaluating the PD-L1 status in nasopharyngeal carcinoma. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 1065–1074. [Google Scholar] [CrossRef]
- Togo, M.; Yokobori, T.; Shimizu, K.; Handa, T.; Kaira, K.; Sano, T.; Tsukagoshi, M.; Higuchi, T.; Yokoo, S.; Shirabe, K.; et al. Diagnostic value of 18F-FDG-PET to predict the tumour immune status defined by tumoural PD-L1 and CD8+ tumour-infiltrating lymphocytes in oral squamous cell carcinoma. Br. J. Cancer 2020, 122, 1686–1694. [Google Scholar] [CrossRef] [Green Version]
- Hirakata, T.; Fujii, T.; Kurozumi, S.; Katayama, A.; Honda, C.; Yanai, K. FDG uptake reflects breast cancer immunological features: The PD-L1 expression and degree of TILs in primary breast cancer. Breast Cancer Res. Treat. 2020, 181, 331–338. [Google Scholar] [CrossRef]
- Seith, F.; Forschner, A.; Schmidt, H.; Pfannenberg, C.; Gückel, B.; Nikolaou, K.; la Fougère, C.; Garbe, C.; Schwenzer, N. 18F-FDG-PET detects complete response to PD-1 therapy in melanoma patients two weeks after therapy start. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 95–101. [Google Scholar] [CrossRef] [PubMed]
- Annovazzi, A.; Vari, S.; Giannarelli, D.; Pasqualoni, R.; Sciuto, R.; Carpano, S.; Cognetti, F.; Ferraresi, V. Comparison of 18F-FDG PET/CT criteria for the prediction of therapy response and clinical outcome in patients with metastatic melanoma treated with ipilimumab and PD-1 inhibitors. Clin. Nucl. Med. 2020, 45, 187–194. [Google Scholar] [CrossRef] [PubMed]
- Tan, A.C.; Emmett, L.; Lo, S.; Liu, V.; Kapoor, R.; Carlino, M.S.; Guminski, A.D.; Long, G.V.; Menzies, A.M. FDG-PET response and outcome from anti-PD-1 therapy in metastatic melanoma. Ann. Oncol. 2018, 29, 2115–2120. [Google Scholar] [CrossRef] [PubMed]
- Humbert, O.; Cadour, N.; Paquet, M.; Schiappa, R.; Poudenx, M.; Chardin, D.; Borchiellini, D.; Benisvy, D.; Ouvrier, M.J.; Zwarthoed, C. 18F-FDG PET/CT in the early assessment of non-small cell lung cancer response to immunotherapy: Frequent and clinical significance of atypical evolutive patterns. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 1158–1167. [Google Scholar] [CrossRef]
- Goldfarb, L.; Duchemann, B.; Chouahnia, K.; Zelek, L.; Soussan, M. Monitoring anti-PD-1 based immunotherapy in non-small cell lung cancer with FDG-PET: Introduction of iPERCIST. EJNMMI Res. 2019, 9, 8. [Google Scholar] [CrossRef]
- Tao, X.; Li, N.; Wu, N.; He, J.; Ying, J.; Gao, S.; Wang, S.; Wang, J.; Wang, Z.; Ling, Y.; et al. The efficiency of 18F-FDG PET-CT for predicting the major pathological response to the neoadjuvant PD-1 blockade in resectable non-small cell lung cancer. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 120919. [Google Scholar] [CrossRef] [Green Version]
- Hashimoto, K.; Kaira, K.; Yamaguchi, O.; Mouri, A.; Shiono, A.; Miura, Y.; Murayama, Y.; Kobayashi, K.; Kagamu, H.; Kuji, I. Potential of FDG-PET as prognostic significance after anti-PD-1 antibody against patients with previously treated non-small cell lung cancer. J. Clin. Med. 2020, 9, 725. [Google Scholar] [CrossRef] [Green Version]
- Seban, R.D.; Mezquita, L.; Berenbaum, A.; Dercle, L.; Botticella, A.; Le Pechoux, C.; Caramella, C.; Deutsch, E.; Grimaldi, S.; Adam, J.; et al. Baseline metabolic tumor burden on FDG PET/CT scans predicts outcome in advanced NSCLC patients treated with immune checkpoint inhibitors. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 1147–1157. [Google Scholar] [CrossRef]
- Takada, K.; Toyokawa, G.; Yoneshima, Y.; Tanaka, K.; Okamoto, I.; Shimokawa, M.; Wakasu, S.; Haro, A.; Osoegawa, A.; Tagawa, T.; et al. 18F-FDG uptake in PET/CT is a potential predictive biomarker of response to anti-PD-1 antibody therapy in non-small cell lung cancer. Sci. Rep. 2019, 9, 13362. [Google Scholar] [CrossRef] [Green Version]
- Chiou, V.L.; Burotto, M. Pseudoprogression and immune-related response in solid tumors. J. Clin. Oncol. 2015, 33, 3541–3543. [Google Scholar] [CrossRef] [Green Version]
- Wolchok, J.D.; Hoos, A.; O’Day, S.; Weber, J.S.; Hamid, O.; Lebbé, C.; Maio, M.; Binder, M.; Bohnsack, O.; Nichol, J.; et al. Guidelines for the evaluation of immune therapy activity in solid tumors: Immune-related response criteria. Clin. Cancer Res. 2009, 15, 7412–7420. [Google Scholar] [CrossRef] [Green Version]
- Nishino, M.; Jagannathan, J.P.; Krajewski, K.M.; O’Regan, K.; Hatabu, H.; Shapiro, G.; Ramaiya, N.H. Personalized tumor response assessment in the era of molecular medicine: Cancer-specific and therapy-specific response criteria to complement pitfalls of RECIST. AJR Am. J. Roentgenol. 2012, 198, 737–745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, S.Y.; Moon, C.M.; Yoon, H.-J.; Kim, B.S.; Lim, J.Y.; Kim, T.O.; Choe, A.R.; Tae, C.H.; Kim, S.E.; Jung, H.K.; et al. Diffuse splenic FDG uptake is predictive of clinical outcomes in patients with rectal cancer. Sci. Rep. 2019, 9, 1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seban, R.-D.; Nemer, J.S.; Marabelle, A.; Yeh, R.; Deutsch, E.; Ammari, S.; Moya-Plana, A.; Mokrane, F.Z.; Gartrell, R.D.; Finkel, G.; et al. Prognostic and theranostic 18F-FDG PET biomarkers for anti-PD1 immunotherapy in metastatic melanoma: Association with outcome and transcriptomics. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 2298–2310. [Google Scholar] [CrossRef] [PubMed]
- De Jaeghere, E.A.; Laloo, F.; Lippens, L.; Van Bockstal, M.; De Man, K.D.; Naert, E.; Van Dorpe, J.; Van de Vijver, K.; Tummers, P.; Makar, A.; et al. Splenic 18F-FDG uptake on baseline PET/CT is associated with oncological outcomes and tumor immune state in uterine cervical cancer. Gynecol. Oncol. 2020, 159, 335–343. [Google Scholar] [CrossRef] [PubMed]
- Seith, F.; Forschner, A.; Weide, B.; Gückel, B.; Schwartz, M.; Schwenck, J.; Othman, A.E.; Fenchel, M.; Garbe, C.; Nikolaou, K.; 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? J. Immunother. Cancer 2020, 8, e000656. [Google Scholar] [CrossRef]
- Dercle, L.; Seban, R.-D.; Lazarovici, J.; Schwartz, L.H.; Houot, R.; Ammari, S.; Danu, A.; Edeline, V.; Marabelle, A.; Ribrag, V.; et al. 18F-FDG PET and CT scans detect new imaging patterns of response and progression in patients with Hodgkin lymphoma treated by anti-programmed death 1 immune checkpoint inhibitor. J. Nucl. Med. 2018, 59, 15–24. [Google Scholar] [CrossRef] [Green Version]
- Ferrari, C.; Maggialetti, N.; Masi, T.; Nappi, A.G.; Santo, G.; Asabella, A.N.; Rubini, G. Early Evaluation of Immunotherapy Response in Lymphoma Patients by 18F-FDG PET/CT: A Literature Overview. J. Pers. Med. 2021, 11, 17. [Google Scholar] [CrossRef]
- Natarajan, A.; Mayer, A.T.; Reeves, R.E.; Nagamine, C.M.; Gambhir, S.S. Development of novel ImmunoPET tracers to image human PD-1 checkpoint expression on tumor-infiltrating lymphocytes in a humanized mouse model. Mol. Imaging Biol. 2017, 19, 903–914. [Google Scholar] [CrossRef] [Green Version]
- Heskamp, S.; Hobo, W.; Molkenboer-Kuenen, J.D.; Olive, D.; Oyen, W.J.G.; Dolstra, H.; Boerman, O.C. Noninvasive imaging of tumor PD-L1 expression using radiolabeled anti-PD-L1 antibodies. Cancer Res. 2015, 75, 2928–2936. [Google Scholar] [CrossRef] [Green Version]
- Larimer, B.M.; Wehrenberg-Klee, E.; Caraballo, A.; Mahmood, U. Quantitative CD3 PET imaging predicts tumor growth response to anti-CTLA-4 therapy. J. Nucl. Med. 2016, 57, 1607–1611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mayer, A.T.; Natarajan, A.; Gordon, S.R.; Maute, R.L.; McCracken, M.N.; Ring, A.M.; Weissman, I.L.; Gambhir, S.S. Practical immuno-PET radiotracer design considerations for human immune checkpoint imaging. J. Nucl. Med. 2017, 58, 538–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maute, R.L.; Gordon, S.R.; Mayer, A.T.; McCracken, M.N.; Natarajan, A.; Ring, N.G.; Kimura, R.; Tsai, J.M.; Manglik, A.; Kruse, A.C.; et al. Engineering high-affinity PD-1 variants for optimized immunotherapy and immuno-PET imaging. Proc. Natl. Acad. Sci. USA 2015, 112, E6506–E6514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Larimer, B.M.; Wehrenberg-Klee, E.; Dubois, F.; Mehta, A.; Kalomeris, T.; Flaherty, K.; Boland, G.; Mahmood, U. Granzyme B PET imaging as a predictive biomarker of immunotherapy response. Cancer Res. 2017, 77, 2318–2327. [Google Scholar] [CrossRef] [Green Version]
- Patel, C.H.; Leone, R.D.; Horton, M.R.; Powell, J.D. Targeting metabolism to regulate immune responses in autoimmunity and cancer. Nat. Rev. Drug Discov. 2019, 18, 669–688. [Google Scholar] [CrossRef]
- Leone, R.D.; Powell, J.D. Metabolism of immune cells in cancer. Nat. Rev. Cancer 2020, 20, 516–531. [Google Scholar] [CrossRef]
- Nguyen, H.A.; Su, Y.; Zhang, J.Y.; Antanasijevic, A.; Caffrey, M.; Schalk, A.M.; Liu, L.; Rondelli, D.; Oh, A.; Mahmud, D.L.; et al. A novel l-asparaginase with low L-glutaminase coactivity is highly efficacious against both T and B cell acute lymphoblastic leukemias in vivo. Cancer Res. 2018, 78, 1549–1560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cruys, B.; Wong, B.W.; Kuchnio, A.; Verdegem, D.; Cantelmo, A.R.; Conradi, L.C.; Vandekeere, S.; Bouché, A.; Cornelissen, I.; Vinckier, S.; et al. Glycolytic regulation of cell rearrangement in angiogenesis. Nat. Commun. 2016, 7, 12240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pavlova, N.N.; Thompson, C.B. The emerging hallmarks of cancer metabolism. Cell Metab. 2016, 23, 27–47. [Google Scholar] [CrossRef] [Green Version]
- Jager, P.L.; Groen, H.J.M.; van der Leest, A.; van Putten, J.W.G.; Pieterman, R.M.; de Vries, E.G.E.; Piers, D.A. L-3-123I-iodo-α-methyl -tyrosine SPECT in non-small cell lung cancer: Preliminary observations. J. Nucl. Med. 2001, 42, 579–585. [Google Scholar]
- Pauleit, D.; Stoffels, G.; Schaden, W.; Hamacher, K.; Bauer, D.; Tellmann, L.; Herzog, H.; Bröer, S.; Coenen, H.H.; Langen, K.J. PET with O-(2-18F fluoroethyl)-L-tyrosine in peripheral tumors: First clinical results. J. Nucl. Med. 2005, 46, 411–416. [Google Scholar] [PubMed]
- Oriuchi, N.; Higuchi, T.; Ishikita, T.; Miyakubo, M.; Hanaoka, H.; Iida, Y.; Endo, K. Present role and future prospect of positron emission tomography in clinical oncology. Cancer Sci. 2006, 97, 1291–1297. [Google Scholar] [CrossRef] [PubMed]
- Tomiyoshi, K.; Inoue, T.; Higuchi, T.; Ahmed, K.; Sarwar, M.; Alyafei, S.; Zhang, H.; Matsubara, K.; Endo, K.; Yang, D. Metabolic studies of [18F-alpha-methyl]tyrosine in mice bearing colorectal carcinoma LS-180. Anti-Cancer Drugs 1999, 10, 329–336. [Google Scholar] [CrossRef]
- Kaira, K.; Oriuchi, N.; Otani, Y.; Shimizu, K.; STanaka, S.; Imai, H.; Yanagitani, N.; Sunaga, N.; Hisada, T.; Ishizuka, T.; et al. Fluorine-18-α-methyltyrosine positron emission tomography for diagnosis and staging of lung cancer: A clinicopathologic study. Clin. Cancer Res. 2007, 13, 6369–6378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, L.; Tominaga, H.; Ohgaki, R.; Wiriyasermkul, P.; Hagiwara, K.; Okuda, S.; Kaira, K.; Oriuchi, N.; Nagamori, S.; Kanai, Y. Specific transport of 3-fluoro-l-α-methyl-tyrosine by LAT1 explains its specificity to malignant tumors in imaging. Cancer Sci. 2016, 107, 347–352. [Google Scholar] [CrossRef]
- Suzuki, S.; Kaira, K.; Ohshima, Y.; Ishioka, N.S.; Sohda, M.; Yokobori, T.; Miyazaki, T.; Oriuchi, N.; Tominaga, H.; Kanai, Y.; et al. Biological significance of fluorine-18-α-methyltyrosine (FAMT) uptake on PET in patients with oesophageal cancer. Br. J. Cancer 2014, 110, 1985–1991. [Google Scholar] [CrossRef] [Green Version]
- Kaira, K.; Higuchi, T.; Sunaga, N.; Arisaka, Y.; Hisada, T.; Tominaga, H.; Oriuchi, N.; Asao, T.; Tsushima, Y.; Yamada, M. Usefulness of 18F-α-methyltyrosine PET for therapeutic monitoring of patients with advanced lung cancer. Anti-Cancer Res. 2016, 36, 6481–6490. [Google Scholar] [CrossRef] [Green Version]
- Shimizu, K.; Kaira, K.; Higuchi, T.; Hisada, T.; Yokobori, T.; Oyama, T.; Asao, T.; Tsushima, Y.; Shirabe, K. Relationship between tumor immune markers and fluorine-18-α-methyltyrosine ([18F]FAMT) uptake in patients with lung cancer. Mol. Imaging Biol. 2020, 22, 1078–1086. [Google Scholar] [CrossRef]
- Häfliger, P.; Charles, R.P. The L-type amino acid transporter LAT1–An emerging target in cancer. Int. J. Mol. Sci. 2019, 20, 2428. [Google Scholar] [CrossRef] [Green Version]
- Okunushi, K.; Furihata, T.; Morio, H.; Muto, Y.; Higuchi, K.; Kaneko, M.; Otsuka, Y.; Ohno, Y.; Watanabe, Y.; Reien, Y.; et al. JPH203, a newly developed anti-cancer drug, shows a preincubation inhibitory effect on L-type amino acid transporter 1 function. J. Pharm. Sci. 2020, 144, 16–22. [Google Scholar] [CrossRef]
- DeBerardinis, R.J.; Chandel, N.S. Fundamentals of cancer metabolism. Sci. Adv. 2016, 2, e1600200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Krall, A.S.; Xu, S.; Graeber, T.G.; Braas, D.; Christofk, H.R. Asparagine promotes cancer cell proliferation through use as an amino acid exchange factor. Nat. Commun. 2016, 7, 11457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Egler, R.A.; Ahuja, S.P.; Matloub, Y. l-Asparaginase in the treatment of patients with acute lymphoblastic leukemia. J. Pharmacol. Pharmacother. 2016, 7, 62–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leone, R.D.; Zhao, L.; Englert, J.M.; Sun, I.M.; Oh, M.H.; Sun, I.M.; Arwood, M.; Bettencourt, I.A.; Patel, C.H.; Wen, J.; et al. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science 2019, 366, 1013–1021. [Google Scholar] [CrossRef]
- Venneti, S.; Dunphy, M.P.; Zhang, H.; Pitter, K.L.; Zanzonico, P.; Campos, C.; Carlin, S.D.; La Rocca, G.; Lyashchenko, S.; Ploessl, K.; et al. Glutamine-based PET imaging facilitates enhanced metabolic evaluation of gliomas in vivo. Sci. Transl. Med. 2015, 7, 274ra17. [Google Scholar] [CrossRef] [Green Version]
- Guo, J.Y.; Teng, X.; Laddha, S.V.; Ma, S.; Van Nostrand, S.C.; Yang, Y.; Khor, S.; Chan, C.S.; Rabinowitz, J.D.; White, E. Autophagy provides metabolic substrates to maintain energy charge and nucleotide pools in Ras-driven lung cancer cells. Genes Dev. 2016, 30, 1704–1717. [Google Scholar] [CrossRef] [Green Version]
- Davidson, S.M.; Jonas, O.; Keibler, M.A.; Hou, H.W.; Luengo, A.; Mayers, J.R.; Wyckoff, J.; Del Rosario, A.M.; Whitman, M.; Chin, C.R.; et al. Direct evidence for cancer cell-autonomous extracellular protein catabolism in pancreatic tumors. Nat. Med. 2017, 23, 235–241. [Google Scholar] [CrossRef]
- Dolgin, E. Anticancer autophagy inhibitors attract ‘resurgent’ interest. Nat. Rev. Drug Discov. 2019, 18, 408–410. [Google Scholar] [CrossRef]
- Karasic, T.B.; O’Hara, M.H.; Loaiza-Bonilla, A.; Reiss, K.A.; Teitelbaum, U.R.; Borazanci, E.; De Jesus-Acosta, A.; Redlinger, C.; Burrell, J.A.; Laheru, D.A.; et al. Effect of gemcitabine and nab-paclitaxel with or without hydroxychloroquine on patients with advanced pancreatic cancer: A phase 2 randomized clinical trial. JAMA Oncol. 2019, 5, 993–998. [Google Scholar] [CrossRef]
- Malhotra, J.; Jabbour, S.; Orlick, M.; Riedlinger, G.; Guo, Y.; White, E.; Aisner, J. Phase Ib/II study of hydroxychloroquine in combination with chemotherapy in patients with metastatic non-small cell lung cancer (NSCLC). Cancer Treat. Res. Commun. 2019, 21, 100158. [Google Scholar] [CrossRef]
- Kudo, Y.; Endo, S.; Fujita, M.; Ota, A.; Kamatari, Y.O.; Tanaka, Y.; Ishikawa, T.; Ikeda, H.; TOkada, T.; Toyooka, N.; et al. Discovery of structure-based optimization of novel Atg4B inhibitors for the treatment of castration-resistant prostate cancer. J. Med. Chem. 2022, 65, 4878–4892. [Google Scholar] [CrossRef] [PubMed]
- Aerts, H.J.; Velazquez, E.R.; Leijenaar, R.T.H.; Parmar, C.; Grossmann, P.; Carvalho, S.; Cavalho, S.; Bussink, J.; Monshouwer, R.; Haibe-Kains, B.; et al. Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach. Nat. Commun. 2014, 5, 4006. [Google Scholar] [CrossRef] [PubMed]
- Zinn, P.O.; Singh, S.K.; Kotrotsou, A.; Hassan, I.; Thomas, G.; Luedi, M.M.; Elakkad, A.; Elshafeey, N.; Idris, T.; Mosley, J.; et al. A co-clinical radiogenomic validation study—Conserved magnetic resonance radiomic appearance of periostin-expressing glioblastoma in patients and xenograft models. Clin. Cancer Res. 2018, 24, 6288–6299. [Google Scholar] [CrossRef] [Green Version]
- Lu, H.; Arshad, M.; Thornton, A.; Avesani, G.; Cunnea, P.; Curry, E.; Kanavati, F.; Liang, J.; Nixon, K.; Williams, S.T.; et al. A mathematical-descriptor of tumor-mesoscopic-structure from computed-tomography images annotates prognostic- and molecular-phenotypes of epithelial ovarian cancer. Nat. Commun. 2019, 10, 764. [Google Scholar] [CrossRef] [PubMed]
- Carles, M.; Fechter, T.; Radicioni, G.; Schimek-Jasch, T.; Adebahr, S.; Zamboglou, C.; Nicolay, N.H.; Martí-Bonmatí, L.; Nestle, U.; Grosu, A.L.; et al. FDG-PET Radiomics for Response Monitoring inNon-Small-Cell Lung Cancer Treated with Radiation Therapy. Cancers 2021, 13, 814. [Google Scholar] [CrossRef]
- Li, H.; Zhang, R.; Wang, S.; Fang, M.; Zhu, Y.; Hu, Z.; Dong, D.; Shi, J.; Tian, J. CT-based radiomic signature as a prognostic factor in stage IV ALK-positive non-small-cell lung cancer treated with TKI crizotinib: A proof-of-concept study. Front. Oncol. 2020, 10, 57. [Google Scholar] [CrossRef] [Green Version]
- Vallières, M.; Zwanenburg, A.; Badic, B.; Le Rest, C.C.; Visvikis, D.; Hatt, M. Responsible Radiomics Research for Faster Clinical Translation. J. Nucl. Med. 2018, 59, 189–194. [Google Scholar] [CrossRef]
- Liu, Z.; Wang, S.; Dong, D.; Wei, J.; Fang, C.; Zhou, X.; Sun, K.; Li, L.; Li, B.; Wang, M.; et al. The Applications of Radiomics in Precision Diagnosis and Treatment of Oncology: Opportunities and Challenges. Theranostics 2019, 9, 1303–1322. [Google Scholar] [CrossRef]
- Oikonomou, A.; Khalvati, F.; Tyrrell, P.N.; Haider, M.A.; Tarique, U.; Jimenez-Juan, L.; Tjong, M.C.; Poon, I.; Eilaghi, A.; Ehrlich, L.; et al. Radiomics analysis at PET/CT contributes to prognosis of recurrence and survival in lung cancer treated with stereotactic body radiotherapy. Sci. Rep. 2018, 8, 4003. [Google Scholar] [CrossRef]
- Aerts, H.J. The potential of radiomic-based phenotyping in precision medicine: A review. JAMA Oncol. 2016, 2, 1636–1642. [Google Scholar] [CrossRef]
- Tixier, F.; Cheze Le Rest, C.; Hatt, M.; Albarghach, N.; Pradier, O.; Metges, J.P.; Corcos, L.; Visvikis, D. Intratumor Heterogeneity Characterized by Textural Features on Baseline 18F-FDG PET Images Predicts Response to Concomitant Radiochemotherapy in Esophageal Cancer. J. Nucl. Med. 2011, 52, 369–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, R.; Limkin, E.J.; Vakalopoulou, M.; Dercle, L.; Champiat, S.; Han, S.R.; Verlingue, L.; Brandao, D.; Lancia, A.; Ammari, S.; et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti- PD-L1 immunotherapy: An imaging biomarker, retrospective multicohort study. Lancet Oncol. 2018, 19, 1180–1191. [Google Scholar] [CrossRef]
- Farwell, M.D.; Gamache, R.F.; Babazada, H.; Hellmann, M.D.; Harding, J.J.; Korn, R.; Mascioni, A.; Le, W.; Wilson, I.; Gordon, M.S.; et al. CD8-Targeted PET Imaging of Tumor-Infiltrating T Cells in Patients with Cancer: A Phase I First-in-Humans Study of 89Zr-Df-IAB22M2C, a Radiolabeled Anti-CD8 Minibody. J. Nucl. Med. 2022, 63, 720–726. [Google Scholar] [CrossRef] [PubMed]
- Kirienko, M.; Sollini, M.; Corbetta, M.; Voulaz, E.; Gozzi, N.; Interlenghi, M.; Gallivanone, F.; Castiglioni, I.; Asselta, R.; Duga, S.; et al. Radiomics and gene expression profile to characterise the disease and predict outcome in patients with lung cancer. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 3643–3655. [Google Scholar] [CrossRef]
- Lv, L.; Xin, B.; Hao, Y.; Yang, Z.; Xu, J.; Wang, L.; Wang, X.; Song, S.; Guo, X. Radiomic analysis for predicting prognosis of colorectal cancer from preoperative 18F-FDG PET/CT. J. Transl. Med. 2022, 20, 66. [Google Scholar] [CrossRef]
- Eertink, J.J.; van de Brug, T.; Wiegers, S.E.; Zwezerijnen, G.J.C.; Pfaehler, E.A.G.; Lugtenburg, P.J.; van der Holt, B.; de Vet, H.C.W.; Hoekstra, O.S.; Boellaard, R.; et al. 18F-FDG PET baseline radiomics features improve the prediction of treatment outcome in diffuse large B-cell lymphoma. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 932–942. [Google Scholar] [CrossRef]
- Sanduleanu, S.; Woodruff, H.C.; de Jong, E.E.C.; van Timmeren, J.E.; Jochems, A.; Dubois, L.; Lambin, P. Tracking tumor biology with radiomics: A systematic review utilizing a radiomics quality score. Radiother. Oncol. 2018, 127, 349–360. [Google Scholar] [CrossRef]
- O’Connor, J.P.B.; Aboagye, E.O.; Adams, J.E.; Aerts, H.J.W.L.; Barrington, S.F.; Beer, A.J.; Boellaard, R.; Bohndiek, S.E.; Brady, M.; Brown, G.; et al. Imaging biomarker roadmap for cancer studies. Nat. Rev. Clin. Oncol. 2017, 14, 169–186. [Google Scholar] [CrossRef]
- Zwanenburg, A.; Vallieres, M.; Abdalah, M.A.; Aerts, H.J.W.L.; Andrearczyk, V.; Apte, A.; Ashrafinia, A.; Bakas, S.; Beukinga, R.J.; Boellaard, R.; et al. The image biomarker standardization initiative: Standardized quantitative radiomics for highthroughput image-based phenotyping. Radiology 2020, 295, 328–338. [Google Scholar] [CrossRef] [Green Version]
- Jha, A.K.; Bradshaw, T.J.; Buvat, I.; Hatt, M.; Kc, P.; Liu, C.; Obuchowski, N.F.; Saboury, B.; Slomka, P.J.; Sunderland, J.J.; et al. Nuclear Medicine and Artificial Intelligence: Best Practices for Evaluation (the RELAINCE guidelines). J. Nucl. Med. 2022, 63, 263239. [Google Scholar] [CrossRef]
- Amyar, A.; Ruan, S.; Gardin, I.; Chatelain, C.; Decazes, P.; Modzelewski, R. 3-D RPET-NET: Development of a 3-D PET imaging convolutional neural network for radiomics analysis and outcome prediction. IEEE Trans. Radiat. Plasma Med Sci. 2019, 3, 225–231. [Google Scholar] [CrossRef]
- Kolinger, G.D.; Garcia, D.V.; Kramer, G.M.; Frings, V.; Zwezerijnen, G.J.C.; Smit, E.F.; de Langen, A.J.; Buvat, I.; Boellaard, R. Effects of Tracer Uptake Time in Non–Small Cell Lung Cancer 18F-FDG PET Radiomics. J. Nucl. Med. 2022, 63, 919–924. [Google Scholar] [CrossRef] [PubMed]
- Zwanenburg, A. Radiomics in nuclear medicine: Robustness, reproducibility, standardization, and how to avoid data analysis traps and replication crisis. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 2638–2655. [Google Scholar] [CrossRef] [PubMed]
Criteria | Measurement | CMR/CR | PMR/PR | PMD/PD | Reference |
---|---|---|---|---|---|
RECIST 1.1 | Unidimensional (LD for non-nodal lesions; LPD for LN) | Disappearance of all target lesions < 10 mm for any pathological LN | ≥30% reduction | ≥20% and ≥5 mm increase, new lesion, or non-target PD | [30] |
irRECIST | Unidimensional (LD for non-nodal lesions; LPD for LN) | Disappearance of all target lesions | ≥30% reduction | ≥20% and ≥5 mm increase, or non-target PD | [33] |
iRECIST | Unidimensional (LD for non-nodal lesions; LPD for LN) | Disappearance of all target lesions | ≥ 30% reduction | ≥20% and ≥5 mm increase, or non-target PD, new lesion confirmed at the next assessment | [34] |
EORTC | SUVmax | Complete resolution of FDG uptake in all lesions | >25% reduction in the sum of SUVmax after more than one cycle of treatment | >25% increase in the sum of SUVmax or appearance of new lesions | [31] |
PERCIST | SULpeak | Complete resolution of FDG uptake in all lesions | ≥30% reduction of SULpeak and an absolute drop of 0.8 SULpeak units | >30% increase in SULpeak and an absolute increase of 0.8 SULpeak, or appearance of new lesions | [32] |
imPERCIST | SULpeak | Complete resolution of FDG uptake in all lesions | ≥30% reduction of SULpeak and an absolute drop of 0.8 SULpeak units | >30% increase in SULpeak and an absolute increase of 0.8 SULpeak, or new lesions included in the sum of SULpeak | [35] |
Cancer Type | Histology | No. of Patients | Correlation between FDG Uptake and PD-L1 Expression p-Value (PD-L1 Clone) | Correlation between FDG Uptake and TILs | Reference |
---|---|---|---|---|---|
Lung cancer | SCC/AC/other | 579 | <0.001 (SP142) | NA | [38] |
Lung cancer | SCC | 167 | 0.02 (E1L3N) | Not significant | [39] |
Lung cancer | AC | 315 | 0.01 (E1L3N/38-8) | Not significant | [40] |
Lung cancer | SCC | 84 | 0.035 (28-8) | NA | [41] |
Bladder cancer | UC/SCC/SRC | 63 | 0.032 (NA) | NA | [42] |
Lung cancer | SCLC | 98 | 0.36 (E1L3N) | Significant | [43] |
Lung cancer | SCC/AC | 362 | 0.001 (28-1) | NA | [44] |
Colon cancer | AC | 65 | 0.001 (28-8) | NA | [45] |
Lung cancer | SCC/AC | 122 | 0.012 (NA) | Significant | [46] |
NPC | SCC | 84 | <0.001 (SP263) | NA | [47] |
OSCC | SCC | 59 | 0.003 (28/8) | Not significant | [48] |
Breast cancer | AC | 97 | <0.001 (28-8) | Significant | [49] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Oriuchi, N.; Endoh, H.; Kaira, K. Monitoring of Current Cancer Therapy by Positron Emission Tomography and Possible Role of Radiomics Assessment. Int. J. Mol. Sci. 2022, 23, 9394. https://doi.org/10.3390/ijms23169394
Oriuchi N, Endoh H, Kaira K. Monitoring of Current Cancer Therapy by Positron Emission Tomography and Possible Role of Radiomics Assessment. International Journal of Molecular Sciences. 2022; 23(16):9394. https://doi.org/10.3390/ijms23169394
Chicago/Turabian StyleOriuchi, Noboru, Hideki Endoh, and Kyoichi Kaira. 2022. "Monitoring of Current Cancer Therapy by Positron Emission Tomography and Possible Role of Radiomics Assessment" International Journal of Molecular Sciences 23, no. 16: 9394. https://doi.org/10.3390/ijms23169394
APA StyleOriuchi, N., Endoh, H., & Kaira, K. (2022). Monitoring of Current Cancer Therapy by Positron Emission Tomography and Possible Role of Radiomics Assessment. International Journal of Molecular Sciences, 23(16), 9394. https://doi.org/10.3390/ijms23169394