PET/CT in Immune-Checkpoint Inhibitor Therapy: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Others
Contributor: , , , , , ,

In this narrative review, experts in nuclear medicine, thoracic oncology, dermatooncology, hemato- and internal oncology, urological and head/neck tumors performed literature reviews in their respective field and a joint discussion on the use of PET/CT in the context of ICI treatment. The aims were to give a clinical overview on present standards and evidence, to identify current challenges and fields of research and to enable an outlook to future developments and their possible implications. 

  • immune checkpoint inhibitor
  • non-small cell lung cancer
  • melanoma
  • lymphoma
  • head and neck cancer
  • renal cancer
  • immunotherapy-related adverse event
  • pseudoprogression
  • tracer
  • CAR-T

Note:All the information in this draft can be edited by authors. And the entry will be online only after authors edit and submit it.

1. Introduction

Positron emission tomography/computed tomography (PET/CT) constitutes a major progress in oncology imaging, as it augments CT with the additional dimension of metabolic activity. Primarily used in staging and to some extent in response assessment of various malignancies, research for additional applications of PET/CT is currently evolving towards prognosis estimation and prediction of response to certain therapies, especially in the field of immunotherapy [1].

Cancer immunotherapy through immune-checkpoint inhibitors (ICI) has revolutionized the world of medical oncology, achieving major and long-term treatment responses also in metastatic disease that would have been unthinkable only a few years ago. Fields of application of ICI therapies have rapidly expanded to different tumor entities and a multitude of ICI substances in various treatment regimens has subsequently become available [2]. Still, by far, not every patient responds to ICI therapies, and existing biomarkers do not allow to predict response precisely on an individual patient’s level. Thus, prediction and monitoring of response to ICI treatment has become a major research target in the recent years.

2. PET/CT in Immune-Checkpoint Inhibitor Therapy—Technical Aspects, Application and Research

2.1. Technical Aspects of PET/CT in Assessing Immunotherapy Response

The advent of ICI in the treatment of various malignant tumors constitutes a milestone in clinical oncology. However, the complex immunological response to these novel agents involving the tumor microenvironment has only been incompletely understood and poses a major challenge in molecular imaging strategies.

Accurate assessment of response to antineoplastic therapy is essential to recognize treatment failure at an early stage and to be able to adapt therapy timely. Classically, this would be indicated by an increase in number and/or size of tumor lesions [3]. Using positron emission tomography (PET), also the change in 18F-FDG uptake in malignant lesions as quantified by the standardized uptake value (SUV) can be used as a biomarker for therapy response. This metabolic biomarker dimension has been included in many oncological treatment concepts. A summary of quantitative biomarkers derived from PET/CT imaging that can be easily (semi)-automatically determined is shown in Table 1.

Table 1. Summary of quantitative biomarkers that can be derived from PET/CT imaging.

SUV (standardized uptake value)

Quantitatively describes the glucose metabolism of a lesion. Regional radioactivity concentrations, determined by the dose administered, the decay of the nuclide and patient’s weight.

SUVmax (maximum standardized uptake value)

Represents the most intensive 18F-FDG uptake in the tumor, maximum SUV value of a region based on a single voxel value only. Often used as a parameter for nuclide uptake, but may be misleading, as it represents only a single voxel value. Thus, it is susceptible to noise, dependent on image resolution, and on the voxel of interest (VOI) definition [4]. An advantage of SUVmax is that placement of the VOI is not critical.

SULpeak (standardized uptake value corrected for lean body mass)

Measured in a 1 cm3 volume around the hottest voxel in the tumor. Is considered a more stable alternative to the noise-susceptible measurement of the SUVmax [5].

MTV (metabolic tumor volume) *

Represents the volume of a tumor lesion with increased 18F-FDG uptake. Whole-body (wb) or total (T) MTV has been defined as the sum of the individual MTVs of all lesions with SUV ≥ 2.5 [6] and has been shown to be a particularly strong prognostic factor in pre-ICI treatment melanoma and NSCLC patients [6–10]. Concerning early response assessment in NSCLC, the increase in wbMTV six weeks after ICI initiation indicated poorer outcomes even in the case of stable disease by CT assessment [11].

TLG (total lesion glycolysis) *

Defined as the product of the MTV and the mean SUV, integrating the tumor-related metabolic activity and tumor volume. In contrast to the SUV, it does not describe the maximum or average glucose turnover at a specific point, but rather the glucose turnover of all lesions. Metabolic tumor response as assessed by TLG may be a more precise predictor of prognosis than MTV or SUVmax [12].

* In contrast to the estimation of SUVmax, determination of MTV and TLG depends on the placement of the VOI. Several approaches can be applied and need to be specified in the methods of the respective publication. Another practical problem with MTV and TLG is that it can be difficult or even impossible to apply in the case of a large number of metastatic lesions, since a VOI has to be created for each individual lesion. ICI: immune-checkpoint inhibitors; NSCLC: non-small-cell lung cancer.

In the context of ICI therapy, however, the efficacy of quantitative measurement of 18F-FDG uptake may be diminished and sometimes misleading. Enhanced 18F-FDG uptake can also be triggered by the activation of the tumor microenvironment with an increased influx and activity of immune cells like T-lymphocytes induced by ICI therapy itself [13,14]. In contrast to aerobic glycolysis in highly differentiated tissues, the so-called Warburg effect in more proliferative neoplastic tissues leads to an increase in anaerobic glycolysis, and thus to an increase in general glucose turnover [15]. At the same time, during immunotherapy, anti-PD-1 activation also stimulates the tumor microenvironment and consequently upregulates glucose transporter (GLUT) mRNA and GLUT proteins, leading to increased glucose consumption as a result of the immunological anti-tumor reaction [16,17]. This altered metabolic situation can conceal the actual treatment response and, under certain circumstances, even lead to false positive scan results. Therefore, new approaches to PET/CT assessment in patients receiving ICI therapy are required [18].

2.2. Standardizing Response-Assessment in PET/CT Imaging

Basic evaluation of therapy response using PET/CT can be accomplished using qualitative parameters, like the decrease or increase in metabolic of active tumor lesions. Such binary “good or poor” categorization is rather robust and can be used, e.g., for end-of-treatment assessment. Particularly complete metabolic response in PET represents an important individual decision-making criterion and generally indicates a favorable long-term outcome [19]. An example of such positive sustained response is shown in Figure 1. Similarly, the appearance of new lesions in follow-up is of higher clinical relevance than changes in preexisting lesions [20].

Figure 1. Complete metabolic response over two years visualized by PET/CT: 68-year-old woman with stage IV lung squamous-cell carcinoma (PD-L1 1%) having progressed after first-line chemotherapy, showing multiple liver, bone and lymph node metastases (a). The patient responded well to pembrolizumab monotherapy, with a PET/CT follow-up scan after one year (b) showing complete metabolic remission. Therapy was completed after two years, still in complete metabolic remission (c). PET/CT: positron emission tomography/computed tomography; PD-L1: programmed death-ligand.

However, an such approach does not allow a fine-tuned therapy assessment and is not suitable for clinical trials, where reproducible and standardized response data are warranted. In conventional CT, this led to the introduction of standardized response evaluation criteria in solid tumors (RECIST) using the classifications complete/partial remission (CR/PR), stable disease (SD) and progressive disease (PD) [21]. Concerning PET/CT, analogous criteria have been suggested, such as the “European Organization for Research and Treatment of Cancer (EORTC) PET Criteria” or more recently the “PET response criteria in solid tumors” (PERCIST) [5,22]. The aim of these assessment tools is to improve the clinical value of metabolic therapy assessment in terms of accuracy, reproducibility and to enable earlier response prediction as compared to conventional imaging techniques, e.g., by CT using size parameters [5].

One major point that needs to be considered in that context is that quantitative indices (including textural analysis) are dependent on the quality of the PET images. Results may vary distinctly between up-to-date imaging technology and equipment ten or twenty years old. Also, comparability is usually impaired when different reconstruction methodologies are used. Both can pose difficulties especially when comparing quantitative measures between institutions [23,24].

2.3. PET/CT and Atypical Response Patterns in Immune-Checkpoint Inhibitor Therapy

In ICI therapy, atypical response patterns pose additional challenges to classical radiological as well as PET-based response evaluations.

One such feature is pseudoprogression [25], which mimics disease progression, although it actually represents a hypermetabolic “flare-phenomenon” caused by the initial T-cell tumor infiltration. Frequently, such patients witness an objective response in the further course of therapy. Pseudoprogression is found considerably less frequently than effective disease progression, with reported rates below 10% [18,25].

Dissociated response is a similar atypical response pattern, with a mixture of lesions responding and progressing simultaneously [26,27]. Its reported frequency varies but might be similar to pseudoprogression, and prognosis is more favorable than for PD [26]; therefore, continuation of ICI treatment should be considered in such patients [27].

Durable responses to ICI therapy are not comprehensively defined but can occur both after primary partial or complete remission or out of stable disease [25]. As shown in Figure 1, 18F-FDG PET/CT can be used to determine long-term remission also on the metabolic level.

An unfavorable feature of atypical response is hyperprogression, which denotes patients presenting with an accelerated tumor growth rate early after ICI initiation, occurring in up to 7% [18,28]. It results in a very poor prognosis and is associated with widespread metastatic disease in the majority of cases [18,29]. Hyperprogression can be easily discovered by CT as well as by 18F-FDG PET-CT, using quantitative parameters, e.g., total lesion glycolysis (TLG) [29].

To account for these imaging challenges associated with atypical response patterns, RECIST criteria have been expanded to iRECIST, which is now commonly used for CT re-staging in ICI patients [30]. Analogously, several similar approaches have been suggested for PET/CT, such as iPERCIST [31], combining RECIST and PERCIST with the introduction of the response category “unconfirmed progressive metabolic disease”. Several other models, such as PERCRIT [32], PERCIMT [13], imPERCIST5 [33] (all for melanoma) or LYRIC (for Hodgkin lymphoma) [34] have been published. However, none of these criteria have been prospectively validated, and all are based on comparably small patient cohorts. Furthermore, differences in timing schedule, reference standards, tumor entities, outcome measures and response classification still limit the practicability of these response criteria in clinical routine.

2.4. PET/CT for Immunotherapy-Related Adverse Events

Immune-related adverse events (IRAE) such as thyreoiditis, pneumonitis or hypophysitis may lead to unusual patterns of 18F-FDG tracer uptake in various involved organs. These therapy-related findings represent possible pitfalls in PET/CT interpretation, especially as such side effects may not necessarily be clinically evident [35–37]. However, they can also have prognostic implications, as their occurrence may indicate a more favorable prognosis [38–40].

2.5. Novel Approaches to PET/CT Imaging and Tracers beyond 18F-FDG:

A novel approach for baseline PET/CT imaging and prognosis estimation apart from traditional staging and response criteria refers to textural features within the tumor lesions. Texture analysis means a systematic, computer-aided evaluation of image data with special regards to features like heterogeneity within one or between individual tumor lesions. Such biomarkers have been correlated with clinical outcome parameters and may pose a major step towards a truly personalized therapy approach. Promising first results have been reported in patients with malignant melanoma treated with vemurafenib and ipilimumab [41]. For lung cancer, similar data could be shown considering the fraction of necrosis within the tumor lesions, which was also linked to the presence of CD8-positive lymphocytes in histological samples [42].

As already discussed, a general problem concerning 18F-FDG as a tracer for cancer treated with ICI is that therapy itself leads to an influx of inflammatory cells leading to enhanced PET activity [43]. Thus, more specific radiopharmaceutical agents are being developed: Radiolabeled ICI like 89Zr-nivolumab or 89Zr-pembrolizumab as well as agents targeting interleukin-2 could aid in the selection of patients who will benefit from ICI-therapy [18]. Data on these substances are still limited to animal models and early-phase human studies; however, an initial trial using 89Zr-atezolizumab for PET imaging showed very promising results assessing clinical response to PD-L1 blockade in 25 patients with locally advanced or metastatic bladder cancer, non-small-cell lung cancer (NSCLC) or triple-negative breast cancer. Interestingly, responses to atezolizumab therapy in these patients had a better correlation with the pretreatment 89Zr-atezolizumab PET signal than with immunohistochemistry- or RNA-sequencing-based predictive markers [44]. Further clinical studies are ongoing and can hopefully be expected in the future.

A very interesting novel approach is based on the use of Zr-89-labeled minibodies that target CD8-positive T cells [45]. Since tumor-infiltrating T cells, in particular CD8-positive T cells, play an important role for initiating and mediating a response to ICI, the in vivo visualization of CD8-positive T-cell-rich tissue might be crucial for development of more effective ICI therapies. A subsequent clinical phase 2 study using 89Zr-IAB22M2C for PET-CT imaging is ongoing among patients with Melanoma, Non-Small-Cell Lung Cancer, Renal Cell Carcinoma, and Squamous Cell Carcinoma of the Head and Neck, aiming to predict response to ICI (NCT03802123).

Another novel promising imaging approach was reported by Chatterjee et al., who developed highly specific radiolabeled peptides for PET imaging of PD-L1 tumor expression [46–48]. Preclinical studies suggest excellent imaging properties and the potential to significantly influence the standard clinical workflow in ICI-treated patients, as these radiopharmaceuticals can be used not only for the therapy guidance and monitoring but also for optimizing dose and therapeutic regimes in a very individual way [48]. Initial clinical trials are ongoing and results may be expected in the near future.

References

  1. Aide, N.; De Pontdeville, M.; Lopci, E. Evaluating response to immunotherapy with 18F-FDG PET/CT: where do we stand? J. Nucl. Med. Mol. Imaging 2020, 47, 1019–1021, doi:10.1007/s00259-020-04702-4.
  2. Wilky, B.A. Immune checkpoint inhibitors: The linchpins of modern immunotherapy. Rev. 2019, 290, 6–23, doi:10.1111/imr.12766.
  3. Therasse, P.; Arbuck, S.G.; Eisenhauer, E.A.; Wanders, J.; Kaplan, R.S.; Rubinstein, L.; Verweij, J.; Van Glabbeke, M.; van Oosterom, A.T.; Christian, M.C.; et al. New Guidelines to Evaluate the Response to Treatment in Solid Tumors. JNCI J. Natl. Cancer Inst. 2000, 92, 205–216, doi:10.1093/jnci/92.3.205.
  4. Boellaard, R.; Krak, N.C.; Hoekstra, O.S.; Lammertsma, A.A. Effects of noise, image resolution, and ROI definition on the accuracy of standard uptake values: a simulation study. Nucl. Med. 2004, 45, 1519–27.
  5. Wahl, R.L.; Jacene, H.; Kasamon, Y.; Lodge, M.A. From RECIST to PERCIST: Evolving Considerations for PET Response Criteria in Solid Tumors. Nucl. Med. 2009, 50, 122S-150S, doi:10.2967/jnumed.108.057307.
  6. Nakamoto, R.; Zaba, L.C.; Rosenberg, J.; Reddy, S.A.; Nobashi, T.W.; Davidzon, G.; Aparici, C.M.; Nguyen, J.; Moradi, F.; Iagaru, A.; et al. Prognostic value of volumetric PET parameters at early response evaluation in melanoma patients treated with immunotherapy. J. Nucl. Med. Mol. Imaging 2020, doi:10.1007/s00259-020-04792-0.
  7. Winther-Larsen, A.; Fledelius, J.; Sorensen, B.S.; Meldgaard, P. Metabolic tumor burden as marker of outcome in advanced EGFR wild-type NSCLC patients treated with erlotinib. Lung Cancer 2016, 94, 81–87, doi:10.1016/j.lungcan.2016.01.024.
  8. 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. J. Nucl. Med. Mol. Imaging 2019, 46, 2298–2310, doi:10.1007/s00259-019-04411-7.
  9. Ito, K.; Schöder, H.; Teng, R.; Humm, J.L.; Ni, A.; Wolchok, J.D.; Weber, W.A. Prognostic value of baseline metabolic tumor volume measured on 18F-fluorodeoxyglucose positron emission tomography/computed tomography in melanoma patients treated with ipilimumab therapy. J. Nucl. Med. Mol. Imaging 2019, 46, 930–939, doi:10.1007/s00259-018-4211-0.
  10. 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. J. Nucl. Med. Mol. Imaging 2020, 47, 1147–1157, doi:10.1007/s00259-019-04615-x.
  11. Spigel, D.R.; Chaft, J.E.; Gettinger, S.; Chao, B.H.; Dirix, L.; Schmid, P.; Chow, L.Q.M.; Hicks, R.J.; Leon, L.; Fredrickson, J.; et al. FIR: Efficacy, Safety, and Biomarker Analysis of a Phase II Open-Label Study of Atezolizumab in PD-L1–Selected Patients With NSCLC. Thorac. Oncol. 2018, 13, 1733–1742, doi:10.1016/j.jtho.2018.05.004.
  12. 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. J. Nucl. Med. Mol. Imaging 2018, 45, 56–66, doi:10.1007/s00259-017-3806-1.
  13. Anwar, H.; Sachpekidis, C.; Winkler, J.; Kopp-Schneider, A.; Haberkorn, U.; Hassel, J.C.; Dimitrakopoulou-Strauss, A. Absolute number of new lesions on 18F-FDG PET/CT is more predictive of clinical response than SUV changes in metastatic melanoma patients receiving ipilimumab. J. Nucl. Med. Mol. Imaging 2018, 45, 376–383, doi:10.1007/s00259-017-3870-6.
  14. Somarouthu, B.; Lee, S.I.; Urban, T.; Sadow, C.A.; Harris, G.J.; Kambadakone, A. Immune-related tumour response assessment criteria: a comprehensive review. J. Radiol. 2018, 91, 20170457, doi:10.1259/bjr.20170457.
  15. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science (80-. ). 2009, 324, 1029–1033, doi:10.1126/science.1160809.
  16. Fu, Y. Facilitative glucose transporter gene expression in human lymphocytes, monocytes, and macrophages: a role for GLUT isoforms 1, 3, and 5 in the immune response and foam cell formation. Blood Cells, Mol. Dis. 2004, 32, 182–190, doi:10.1016/j.bcmd.2003.09.002.
  17. Maratou, E.; Dimitriadis, G.; Kollias, A.; Boutati, E.; Lambadiari, V.; Mitrou, P.; Raptis, S.A. Glucose transporter expression on the plasma membrane of resting and activated white blood cells. J. Clin. Invest. 2007, 37, 282–290, doi:10.1111/j.1365-2362.2007.01786.x.
  18. Decazes, P.; Bohn, P. Immunotherapy by Immune Checkpoint Inhibitors and Nuclear Medicine Imaging: Current and Future Applications. Cancers (Basel). 2020, 12, 371, doi:10.3390/cancers12020371.
  19. 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. Oncol. 2018, 29, 2115–2120, doi:10.1093/annonc/mdy330.
  20. Iravani, A.; Hicks, R.J. Imaging the Cancer Immune Environment and Its Response to Pharmacologic Intervention, Part 1: The Role of 18 F-FDG PET/CT. Nucl. Med. 2020, 61, 943–950, doi:10.2967/jnumed.119.234278.
  21. 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). J. Cancer 2009, 45, 228–247, doi:10.1016/j.ejca.2008.10.026.
  22. Pinker, K.; Riedl, C.; Weber, W.A. Evaluating tumor response with FDG PET: updates on PERCIST, comparison with EORTC criteria and clues to future developments. J. Nucl. Med. Mol. Imaging 2017, 44, 55–66, doi:10.1007/s00259-017-3687-3.
  23. Sunderland, J.J.; Christian, P.E. Quantitative PET/CT Scanner Performance Characterization Based Upon the Society of Nuclear Medicine and Molecular Imaging Clinical Trials Network Oncology Clinical Simulator Phantom. Nucl. Med. 2015, 56, 145–152, doi:10.2967/jnumed.114.148056.
  24. Graham, M.M.; Badawi, R.D.; Wahl, R.L. Variations in PET/CT Methodology for Oncologic Imaging at U.S. Academic Medical Centers: An Imaging Response Assessment Team Survey. Nucl. Med. 2011, 52, 311–317, doi:10.2967/jnumed.109.074104.
  25. Borcoman, E.; Kanjanapan, Y.; Champiat, S.; Kato, S.; Servois, V.; Kurzrock, R.; Goel, S.; Bedard, P.; Le Tourneau, C. Novel patterns of response under immunotherapy. Oncol. 2019, 30, 385–396, doi:10.1093/annonc/mdz003.
  26. Tazdait, M.; Mezquita, L.; Lahmar, J.; Ferrara, R.; Bidault, F.; Ammari, S.; Balleyguier, C.; Planchard, D.; Gazzah, A.; Soria, J.C.; et al. Patterns of responses in metastatic NSCLC during PD-1 or PDL-1 inhibitor therapy: Comparison of RECIST 1.1, irRECIST and iRECIST criteria. J. Cancer 2018, 88, 38–47, doi:10.1016/j.ejca.2017.10.017.
  27. Humbert, O.; Cadour, N.; Paquet, M.; Schiappa, R.; Poudenx, M.; Chardin, D.; Borchiellini, D.; Benisvy, D.; Ouvrier, M.J.; Zwarthoed, C.; et al. 18FDG PET/CT in the early assessment of non-small cell lung cancer response to immunotherapy: frequency and clinical significance of atypical evolutive patterns. J. Nucl. Med. Mol. Imaging 2020, 47, 1158–1167, doi:10.1007/s00259-019-04573-4.
  28. Kanjanapan, Y.; Day, D.; Wang, L.; Al‐Sawaihey, H.; Abbas, E.; Namini, A.; Siu, L.L.; Hansen, A.; Razak, A.A.; Spreafico, A.; et al. Hyperprogressive disease in early‐phase immunotherapy trials: Clinical predictors and association with immune‐related toxicities. Cancer 2019, 125, 1341–1349, doi:10.1002/cncr.31999.
  29. Ferrara, R.; Mezquita, L.; Texier, M.; Lahmar, J.; Audigier-Valette, C.; Tessonnier, L.; Mazieres, J.; Zalcman, G.; Brosseau, S.; Le Moulec, S.; et al. Hyperprogressive Disease in Patients With Advanced Non–Small Cell Lung Cancer Treated With PD-1/PD-L1 Inhibitors or With Single-Agent Chemotherapy. JAMA Oncol. 2018, 4, 1543, doi:10.1001/jamaoncol.2018.3676.
  30. 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, doi:10.1016/S1470-2045(17)30074-8.
  31. 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, doi:10.1186/s13550-019-0473-1.
  32. Cho, S.Y.; Lipson, E.J.; Im, H.-J.; Rowe, S.P.; Gonzalez, E.M.; Blackford, A.; Chirindel, A.; Pardoll, D.M.; Topalian, S.L.; Wahl, R.L. Prediction of Response to Immune Checkpoint Inhibitor Therapy Using Early-Time-Point 18 F-FDG PET/CT Imaging in Patients with Advanced Melanoma. Nucl. Med. 2017, 58, 1421–1428, doi:10.2967/jnumed.116.188839.
  33. Ito, K.; Teng, R.; Schöder, H.; Humm, J.L.; Ni, A.; Michaud, L.; Nakajima, R.; Yamashita, R.; Wolchok, J.D.; Weber, W.A. 18 F-FDG PET/CT for Monitoring of Ipilimumab Therapy in Patients with Metastatic Melanoma. Nucl. Med. 2019, 60, 335–341, doi:10.2967/jnumed.118.213652.
  34. Chen, A.; Mokrane, F.-Z.; Schwartz, L.H.; Morschhauser, F.; Stamatoullas, A.; Schiano de Colella, J.-M.; Vercellino, L.; Casasnovas, O.; Chauchet, A.; Delmer, A.; et al. Early 18 F-FDG PET/CT Response Predicts Survival in Relapsed or Refractory Hodgkin Lymphoma Treated with Nivolumab. Nucl. Med. 2020, 61, 649–654, doi:10.2967/jnumed.119.232827.
  35. Aide, N.; Hicks, R.J.; Le Tourneau, C.; Lheureux, S.; Fanti, S.; Lopci, E. FDG PET/CT for assessing tumour response to immunotherapy. J. Nucl. Med. Mol. Imaging 2019, 46, 238–250, doi:10.1007/s00259-018-4171-4.
  36. Martins, F.; Sofiya, L.; Sykiotis, G.P.; Lamine, F.; Maillard, M.; Fraga, M.; Shabafrouz, K.; Ribi, C.; Cairoli, A.; Guex-Crosier, Y.; et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Rev. Clin. Oncol. 2019, 16, 563–580, doi:10.1038/s41571-019-0218-0.
  37. Tirumani, S.H.; Ramaiya, N.H.; Keraliya, A.; Bailey, N.D.; Ott, P.A.; Hodi, F.S.; Nishino, M. Radiographic Profiling of Immune-Related Adverse Events in Advanced Melanoma Patients Treated with Ipilimumab. Cancer Immunol. Res. 2015, 3, 1185–1192, doi:10.1158/2326-6066.CIR-15-0102.
  38. Sachpekidis, C.; Larribère, L.; Kopp-Schneider, A.; Hassel, J.C.; Dimitrakopoulou-Strauss, A. Can benign lymphoid tissue changes in 18F-FDG PET/CT predict response to immunotherapy in metastatic melanoma? Cancer Immunol. Immunother. 2019, 68, 297–303, doi:10.1007/s00262-018-2279-9.
  39. Sachpekidis, C.; Kopp-Schneider, A.; Hakim-Meibodi, L.; Dimitrakopoulou-Strauss, A.; Hassel, J.C. 18F-FDG PET/CT longitudinal studies in patients with advanced metastatic melanoma for response evaluation of combination treatment with vemurafenib and ipilimumab. Melanoma Res. 2019, 29, 178–186, doi:10.1097/CMR.0000000000000541.
  40. Nobashi, T.; Baratto, L.; Reddy, S.A.; Srinivas, S.; Toriihara, A.; Hatami, N.; Yohannan, T.K.; Mittra, E. Predicting Response to Immunotherapy by Evaluating Tumors, Lymphoid Cell-Rich Organs, and Immune-Related Adverse Events Using FDG-PET/CT. Nucl. Med. 2019, 44, e272–e279, doi:10.1097/RLU.0000000000002453.
  41. Dittrich, D.; Pyka, T.; Scheidhauer, K.; Lütje, S.; Essler, M.; Bundschuh, R.A. Textural features in FDG-PET/CT can predict outcome in melanoma patients to treatment with Vemurafenib and Ipililumab. Nuklearmedizin 2020, 59, 228–234, doi:10.1055/a-1140-5458.
  42. Jreige, M.; Letovanec, I.; Chaba, K.; Renaud, S.; Rusakiewicz, S.; Cristina, V.; Peters, S.; Krueger, T.; de Leval, L.; Kandalaft, L.E.; et al. 18F-FDG PET metabolic-to-morphological volume ratio predicts PD-L1 tumour expression and response to PD-1 blockade in non-small-cell lung cancer. J. Nucl. Med. Mol. Imaging 2019, 46, 1859–1868, doi:10.1007/s00259-019-04348-x.
  43. Laura, E.; Sepulcri, M.; Pasello, G. PET/CT And The Response To Immunotherapy In Lung Cancer. Radiopharm. 2019, 13, doi:10.2174/1874471013666191220105449.
  44. Bensch, F.; van der Veen, E.L.; Lub-de Hooge, M.N.; Jorritsma-Smit, A.; Boellaard, R.; Kok, I.C.; Oosting, S.F.; Schröder, C.P.; Hiltermann, T.J.N.; van der Wekken, A.J.; et al. 89Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer. Med. 2018, 24, 1852–1858, doi:10.1038/s41591-018-0255-8.
  45. Pandit-Taskar, N.; Postow, M.A.; Hellmann, M.D.; Harding, J.J.; Barker, C.A.; O’Donoghue, J.A.; Ziolkowska, M.; Ruan, S.; Lyashchenko, S.K.; Tsai, F.; et al. First-in-Humans Imaging with 89 Zr-Df-IAB22M2C Anti-CD8 Minibody in Patients with Solid Malignancies: Preliminary Pharmacokinetics, Biodistribution, and Lesion Targeting. Nucl. Med. 2020, 61, 512–519, doi:10.2967/jnumed.119.229781.
  46. Chatterjee, S.; Lesniak, W.G.; Miller, M.S.; Lisok, A.; Sikorska, E.; Wharram, B.; Kumar, D.; Gabrielson, M.; Pomper, M.G.; Gabelli, S.B.; et al. Rapid PD-L1 detection in tumors with PET using a highly specific peptide. Biophys. Res. Commun. 2017, 483, 258–263, doi:10.1016/j.bbrc.2016.12.156.
  47. Lesniak, W.G.; Mease, R.C.; Chatterjee, S.; Kumar, D.; Lisok, A.; Wharram, B.; Kalagadda, V.R.; Emens, L.A.; Pomper, M.G.; Nimmagadda, S. Development of [ 18 F]FPy-WL12 as a PD-L1 Specific PET Imaging Peptide. Imaging 2019, 18, 153601211985218, doi:10.1177/1536012119852189.
  48. Kumar, D.; Lisok, A.; Dahmane, E.; McCoy, M.; Shelake, S.; Chatterjee, S.; Allaj, V.; Sysa-Shah, P.; Wharram, B.; Lesniak, W.G.; et al. Peptide-based PET quantifies target engagement of PD-L1 therapeutics. Clin. Invest. 2019, 129, 616–630, doi:10.1172/JCI122216.

This entry is adapted from the peer-reviewed paper 10.3390/jcm9113483

This entry is offline, you can click here to edit this entry!
Video Production Service