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?

Ferdinand Seith, Andrea Forschner, Benjamin Weide, Brigitte Gückel, Martin Schwartz, Johannes Schwenck, Ahmed E Othman, Matthias Fenchel, Claus Garbe, Konstantin Nikolaou, Nina Schwenzer, Christian la Fougère, Christina Pfannenberg, Ferdinand Seith, Andrea Forschner, Benjamin Weide, Brigitte Gückel, Martin Schwartz, Johannes Schwenck, Ahmed E Othman, Matthias Fenchel, Claus Garbe, Konstantin Nikolaou, Nina Schwenzer, Christian la Fougère, Christina Pfannenberg

Abstract

Response assessment or prediction to checkpoint inhibitor therapy (CIT) is an unsolved problem in current routine diagnostics of patients with melanoma. Here, we evaluated very early changes of primary and secondary lymphoid organs under CIT in multiparametric [18F]-labeled fluorodeoxyglucose-positron emission tomography (18F-FDG-PET)/MRI as possible predictors of treatment response and investigated their correlation with baseline blood immune biomarkers. Between October 2014 and November 2017, 17 patients with unresectable melanoma (8 females; 65±11 years) undergoing CIT were prospectively evaluated using whole-body 18F-FDG-PET/MRI before CIT start (t0), 2 weeks (t1) and 3 months after CIT initiation (t2). At each time point, the volume, the 18F-FDG-uptake and the mean apparent diffusion coefficient (ADC) of the spleen as well as the 18F-FDG uptake of the bone marrow were assessed. Relative lymphocyte count (RLC), relative eosinophil count (REC) and neutrophil-lymphocyte ratio (NLR) were assessed at baseline. Response Evaluation Criteria in Solid Tumours modified for immune-based therapeutics (iRECIST) and decisions from an interdisciplinary tumor board were used for treatment response evaluation at t2 iRECIST was compared with PET response criteria in solid tumors for image-based response evaluation at different time points. Comparative analysis was conducted with Mann-Whitney U test with false discovery rate correction for multiple testing and correlation coefficients were computed. In lymphoid organs, significant differences (p<0.05) between responders (9/17) and non-responders were found for the 18F-FDG-uptake in the spleen at t1 and the increase of the uptake t1-t0 (responders/non-responders: standardized uptake value lean body mass 1.19/0.93; +49%/-1%). The best correlation coefficients to baseline biomarkers were found for the 18F-FDG-uptake in the spleen at t1: NLR, r=-0.46; RLC, r=0.43; REC, r=0.58 (p<0.05), respectively. Compared with the non-responder group, the responder group showed marked increases also in the volume of the spleen (+22%/+10%), the 18F-FDG-uptake of bone marrow (+31%/-9%) at t1 and the ADCmean at t2 (+46%/+15%) compared with t0, however, not reaching significance. Our findings indicate that an effective systemic immune response in patients undergoing CIT can be detected as a significantly increased spleen activity in 18F-FDG-PET as early as 2 weeks after treatment initiation. TRIAL REGISTRATION NUMBER: NCT03132090, DRKS00013925.

Keywords: CTLA-4 antigen; melanoma; programmed cell death 1 receptor.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Overview of cohort selection. A high number of patients quit the study and changes of therapy during the 3 months of observation resulted in a high drop-out rate. In five patients, no metastases with an increased uptake of [18F]-labeled fluorodeoxyglucose (18F-FDG) were found: in two patients, lesions turned out to be inflammatory (lung and liver), in one patient, a spinal meningeal melanocytoma was finally diagnosed and in two patients, suspicious lesions did not show an increased glucose consumption and were therefore not rated as metastases. CIT, checkpoint inhibitor therapy.
Figure 2
Figure 2
Left hand side: DIXON MRI (in and opposed phase) were used to compute a bone mask of the spine (highlighted in the positron emission tomography (PET) maximum intensity projection image), which was copied to the PET images to assess the standardized uptake value lean body mass (SULmean) of the bone marrow. Right hand side: example of a responder (patient no. 10, also shown in online supplementary figure S2) and a non-responder (patient no. 11) to checkpoint inhibitor therapy. Note the changes of [18F]-labeled fluorodeoxyglucose (18F-FDG)-uptake in the bone marrow in the responder especially at t1 while there was no significant change observed in the non-responder.
Figure 3
Figure 3
Morphological (iRECIST) and metabolic (positron emission tomography (PET) response criteria in solid tumors (PERCIST)) response assessment (upper two rows, ‘Response’) as well as multiparametric changes in the spleen (‘Spleen’) and baseline immune biomarkers (bottom line) in a responder (left hand side, patient no. 14) and a non-responder (right hand side, patient no. 11) to checkpoint inhibitor therapy. Left hand side: metastases in the liver and the lung at t0 (black arrows) with an excellent treatment response already visible at t1 in PET: the metastases in the lung disappears in PET and the metabolic activity of the liver metastases decreases significantly, resulting in partial metabolic response (PMR); diameters of metastases did not show significant changes, leading to immune stable disease (iSD) in iRECIST at t1. Avital tumor residue in the liver at t2 (immune partial response (iPR)) without specific tracer uptake (complete metabolic response (CMR)). Note the significant increase of metabolic activity and the volume of the spleen (dotted ring in the PET/MRI), especially at t1 as compared with t0, considerably less pronounced at t2. Right hand side: metastases in the liver, the lung and the soft tissue of the left leg (black arrows). New metastases are visible in both MRI and PET already at t1, resulting in PMD/immune progressive disease (iPD). The volume of the spleen slightly increases under therapy, but the metabolic activity remains stable. The apparent diffusion coefficient (ADC)mean of the spleen did not show clear trends under therapy. The dotted regions of interes (ROIs) in the images aim to highlight the spleen and are not the volume of interest (VOI) used for image evaluation.
Figure 4
Figure 4
Left hand side: results of the evaluation of lymphoid organs demonstrated as relative changes in % to the baseline scan t0 in the responder and non-responder group. Significant differences between the time points were found for SULmean at t1 and for relative differences between t0 and t1. Right hand side: correlation of metabolic activity of the spleen at t1 (SULmean t1) to the baseline immune biomarkers NLR, RLC and REC of all patients. NLR, neutrophile-lymphocyte ratio; REC, relative eosinophil count; RLC, relative lymphocyte count; SULmean, standardized uptake value lean body mass.

References

    1. Coley WB. The treatment of malignant tumors by repeated inoculations of erysipelas. with a report of ten original cases. 1893. Clin Orthop Relat Res 1991;262:3–11.
    1. Lipson EJ, Forde PM, Hammers H-J, et al. . Antagonists of PD-1 and PD-L1 in cancer treatment. Semin Oncol 2015;42:587–600. 10.1053/j.seminoncol.2015.05.013
    1. Brahmer J, Reckamp KL, Baas P, et al. . Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med 2015;373:123–35. 10.1056/NEJMoa1504627
    1. Pflugfelder A, Kochs C, Blum A, et al. . Malignant Melanoma S3-Guideline “Diagnosis, Therapy and Follow-up of Melanoma”. J Dtsch Dermatol Ges 2013;11:1–116. 10.1111/ddg.12113_suppl
    1. Xu-Monette ZY, Zhou J, Young KH. Pd-1 expression and clinical PD-1 blockade in B-cell lymphomas. Blood 2018;131:68–83. 10.1182/blood-2017-07-740993
    1. Wolchok JD, Kluger H, Callahan MK, et al. . Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013;369:122–33. 10.1056/NEJMoa1302369
    1. Hamid O, Robert C, Daud A, et al. . Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 2013;369:134–44. 10.1056/NEJMoa1305133
    1. Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature 2011;480:480–9. 10.1038/nature10673
    1. Ribas A, Hamid O, Daud A, et al. . Association of pembrolizumab with tumor response and survival among patients with advanced melanoma. JAMA 2016;315:1600–9. 10.1001/jama.2016.4059
    1. Robert C, Schachter J, Long GV, et al. . Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 2015;372:2521–32. 10.1056/NEJMoa1503093
    1. Ribas A, Puzanov I, Dummer R, et al. . Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol 2015;16:908–18. 10.1016/S1470-2045(15)00083-2
    1. Hofmann L, Forschner A, Loquai C, et al. . Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of anti-PD-1 therapy. Eur J Cancer 2016;60:190–209. 10.1016/j.ejca.2016.02.025
    1. Champiat S, Dercle L, Ammari S, et al. . Hyperprogressive disease is a new pattern of progression in cancer patients treated by anti-PD-1/PD-L1. Clin Cancer Res 2017;23:1920–8. 10.1158/1078-0432.CCR-16-1741
    1. Wolchok JD, Hoos A, O'Day S, et al. . Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res 2009;15:7412–20. 10.1158/1078-0432.CCR-09-1624
    1. Sachpekidis C, Anwar H, Winkler J, et al. . The role of interim 18F-FDG PET/CT in prediction of response to ipilimumab treatment in metastatic melanoma. Eur J Nucl Med Mol Imaging 2018;45:1289–96. 10.1007/s00259-018-3972-9
    1. Seith F, Forschner A, Schmidt H, et al. . 18F-Fdg-Pet detects complete response to PD1-therapy in melanoma patients two weeks after therapy start. Eur J Nucl Med Mol Imaging 2018;45:95–101. 10.1007/s00259-017-3813-2
    1. Spitzer MH, Carmi Y, Reticker-Flynn NE, et al. . Systemic immunity is required for effective cancer immunotherapy. Cell 2017;168:e15:487–502. 10.1016/j.cell.2016.12.022
    1. Jacquelot N, Roberti MP, Enot DP, et al. . Predictors of responses to immune checkpoint blockade in advanced melanoma. Nat Commun 2017;8:592. 10.1038/s41467-017-00608-2
    1. Martens A, Wistuba-Hamprecht K, Geukes Foppen M, et al. . Baseline peripheral blood biomarkers associated with clinical outcome of advanced melanoma patients treated with ipilimumab. Clin Cancer Res 2016;22:2908–18. 10.1158/1078-0432.CCR-15-2412
    1. Capone M, Giannarelli D, Mallardo D, et al. . Baseline neutrophil-to-lymphocyte ratio (NLR) and derived NLR could predict overall survival in patients with advanced melanoma treated with nivolumab. J Immunother Cancer 2018;6:74. 10.1186/s40425-018-0383-1
    1. Lino-Silva LS, Salcedo-Hernández RA, García-Pérez L, et al. . Basal neutrophil-to-lymphocyte ratio is associated with overall survival in melanoma. Melanoma Res 2017;27:140–4. 10.1097/CMR.0000000000000333
    1. Schwenck J, Schörg B, Fiz F, et al. . Cancer immunotherapy is accompanied by distinct metabolic patterns in primary and secondary lymphoid organs observed by non-invasive in vivo18F-FDG-PET. Theranostics 2020;10:925–37. 10.7150/thno.35989
    1. Radu CG, Shu CJ, Nair-Gill E, et al. . Molecular imaging of lymphoid organs and immune activation by positron emission tomography with a new [18F]-labeled 2'-deoxycytidine analog. Nat Med 2008;14:783–8. 10.1038/nm1724
    1. Padhani AR, Koh D-M, Collins DJ. Whole-Body diffusion-weighted MR imaging in cancer: current status and research directions. Radiology 2011;261:700–18. 10.1148/radiol.11110474
    1. Wahl RL, Jacene H, Kasamon Y, et al. . From RECIST to PERCIST: evolving considerations for PET response criteria in solid tumors. J Nucl Med 2009;50 Suppl 1:122S–50. 10.2967/jnumed.108.057307
    1. Seymour L, Bogaerts J, Perrone A, et al. . iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol 2017;18:e143–52. 10.1016/S1470-2045(17)30074-8
    1. Kaira K, Higuchi T, Naruse I, 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. 10.1007/s00259-017-3806-1
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. . New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228–47. 10.1016/j.ejca.2008.10.026
    1. Cho SY, Lipson EJ, Im H-J, et al. . Prediction of Response to Immune Checkpoint Inhibitor Therapy Using Early-Time-Point 18F-FDG PET/CT Imaging in Patients with Advanced Melanoma. J Nucl Med 2017;58:1421–8. 10.2967/jnumed.116.188839
    1. Anwar H, Sachpekidis C, Winkler J, et al. . 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. Eur J Nucl Med Mol Imaging 2018;45:376–83. 10.1007/s00259-017-3870-6
    1. Dercle L, Seban R-D, Lazarovici J, 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. 10.2967/jnumed.117.193011
    1. Tsai KK, Pampaloni MH, Hope C, et al. . Increased FDG avidity in lymphoid tissue associated with response to combined immune checkpoint blockade. J Immunother Cancer 2016;4:58. 10.1186/s40425-016-0162-9
    1. Aide N, Hicks RJ, Le Tourneau C, et al. . FDG PET/CT for assessing tumour response to immunotherapy : Report on the EANM symposium on immune modulation and recent review of the literature. Eur J Nucl Med Mol Imaging 2019;46:238–50. 10.1007/s00259-018-4171-4
    1. Tavaré R, Escuin-Ordinas H, Mok S, et al. . An effective Immuno-PET imaging method to monitor CD8-Dependent responses to immunotherapy. Cancer Res 2016;76:73–82. 10.1158/0008-5472.CAN-15-1707
    1. Racanelli V, Rehermann B. The liver as an immunological organ. Hepatology 2006;43:S54–62. 10.1002/hep.21060
    1. Gkiozos I, Kopitopoulou A, Kalkanis A, et al. . Sarcoidosis-Like reactions induced by checkpoint inhibitors. J Thorac Oncol 2018;13:1076–82. 10.1016/j.jtho.2018.04.031
    1. Gückel B, Gatidis S, Enck P, et al. . Patient comfort during positron emission tomography/magnetic resonance and positron emission tomography/computed tomography examinations: subjective assessments with visual analog scales. Invest Radiol 2015;50:726–32. 10.1097/RLI.0000000000000177
    1. Taron J, Schraml C, Pfannenberg C, et al. . Simultaneous multislice diffusion-weighted imaging in whole-body positron emission tomography/magnetic resonance imaging for multiparametric examination in oncological patients. Eur Radiol 2018;28:3372–83. 10.1007/s00330-017-5216-y

Source: PubMed

3
Subscribe