FLT PET/CT imaging of metastatic prostate cancer patients treated with pTVG-HP DNA vaccine and pembrolizumab

Matthew Scarpelli, Christopher Zahm, Scott Perlman, Douglas G McNeel, Robert Jeraj, Glenn Liu, Matthew Scarpelli, Christopher Zahm, Scott Perlman, Douglas G McNeel, Robert Jeraj, Glenn Liu

Abstract

Background: Immunotherapy has demonstrated remarkable success in treating different cancers. Nonetheless, a large number of patients do not respond, many respond without immediate changes detectable with conventional imaging, and many have unusual immune-related adverse events that cannot be predicted in advance. In this exploratory study, we investigate how 3'-Deoxy-3'-18F-fluorothymidine (FLT) positron emission tomography (PET) measurements of tumor and immune cell proliferation might be utilized as biomarkers in immunotherapy.

Methods: Seventeen patients with metastatic castrate resistant prostate cancer were treated with combination pTVG-HP DNA vaccine and pembrolizumab. Patients underwent baseline and 12-week FLT PET/CT scans. FLT PET standardized uptake values (SUVs) were extracted from tumors, non-metastatic lymph nodes, spleen, bone marrow, pancreas, and thyroid to quantify cell proliferation in these tissues. Regional immune cell responses to pTVG-HP DNA vaccine were assessed by comparing FLT uptake changes in vaccine draining and non-draining lymph nodes. Cox proportional hazards regression was utilized to relate FLT uptake and other clinical markers (PSA and tumor size) to progression-free survival. Area under receiver operating characteristic (AUC) curves and concordance indices were used to assess the predictive capabilities of FLT uptake.

Results: Changes in FLT uptake in vaccine draining lymph nodes were significantly greater than changes in non-draining lymph nodes (P = 0.02), suggesting a regional immune response to vaccination. However, the changes in FLT uptake in lymph nodes were not significantly predictive of progression-free survival. Increases in tumor FLT uptake were significantly predictive of shorter progression-free survival (concordance index = 0.83, P < 0.01). Baseline FLT uptake in the thyroid was significantly predictive of whether or not a patient would subsequently experience a thyroid-related adverse event (AUC = 0.97, P < 0.01).

Conclusions: FLT PET uptake was significantly predictive of progression-free survival and the occurrence of adverse events relating to thyroid function. The results suggest FLT PET imaging has potential as a biomarker in immunotherapy, providing a marker of tumor and immune responses, and as a possible means of anticipating specific immune-related adverse events.

Trial registration: NCT02499835 .

Keywords: Adverse events; Cell proliferation; Clinical trial; DNA vaccine; FLT PET; Imaging; Pembrolizumab; Prostate cancer; Response assessment.

Conflict of interest statement

Ethics approval and consent to participate

The protocol for this study was reviewed and approved by all local (University of Wisconsin Human Subjects’ Review Board (IRB). All patients gave written informed consent for participation.

Consent for publication

Not applicable.

Competing interests

Douglas G. McNeel has ownership interest, has received research support, and serves as consultant to Madison Vaccines, Inc. which has licensed intellectual property related to this content. None of the other authors have relevant potential conflicts of interest.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Patients are numbered the same in all inserts (also the same as in Table 1) and are ordered by study arm. a Changes in FLT SUVmean in vaccine draining left axillary lymph nodes are shown for each patient along with changes in non-draining right axillary lymph nodes. A number of outliers with changes in left axillary lymph node SUVmean greater than 50% are evident. b Representative FLT PET/CT slice is showing a vaccine draining left axillary lymph with elevated uptake after 12 weeks of therapy (patient #1) c Changes in SUVmean in bone marrow and spleen. d Representative FLT PET/CT slice is showing increased splenic FLT uptake after 12 weeks (patient #17). e Changes in FLT uptake in patients with soft tissue metastases are shown for SUVmean and SUVtotal. Changes in tumor SUVmean and tumor SUVtotal were significantly correlated (ρ = 0.66, P = 0.04). f Representative FLT PET/CT slice is showing metastatic mediastinal lymph nodes with visually increased FLT uptake after 12 weeks of therapy (patient #9). Following RECIST guidelines this patient had radiographically stable disease at week 12 but had subsequent disease progression upon the next radiographic follow-up at 24 weeks. g Changes in thyroid FLT uptake h Representative PET/CT slice is shown for a patient that experienced grade 2 hyperthyroidism (patient #11). The arrow indicates the position of the right thyroid lobe where visually increased FLT uptake is evident at 12 weeks. Notably, this patient had their first pembrolizumab injection 1 day prior to their 12-week PET scan
Fig. 2
Fig. 2
a Changes in tumor FLT SUVmean after 12 weeks are plotted against changes in tumor size after 12 weeks. Tumor size was measured following RECIST guidelines using a diagnostic CT scan. b Changes in tumor FLT SUVmean after 12 weeks are plotted against changes in tumor size after 24 weeks. c Changes in tumor FLT SUVmean after 12 weeks are plotted against changes in PSA after 12 weeks. d Changes in tumor FLT SUVmean after 12 weeks are plotted against changes in PSA after 24 weeks. Note some patients are not included in these figures because they did not have RECIST measurable soft tissue tumors or were on study for less than 24 weeks
Fig. 3
Fig. 3
a Change in tumor SUVmean at 12 weeks differentiated patients who had progression-free survival less than or equal to the median progression-free survival time (24 weeks) from patients who had progression-free survival greater than the median. b Changes in PSA levels after 12 weeks for the same set of patients as shown in insert (a)
Fig. 4
Fig. 4
a Axial CT and PET/CT slices with a metastatic tumor indicated. At week 12 this patient had experienced diminished PSA and RECIST measurements but increased tumor FLT uptake. By week 16, this patient was found to have progressive disease with marked increases in tumor size and PSA. b Immunofluorescence images show representative FFPE sections taken from the week 12 biopsy of the tumor indicated in part (a). The left immunofluorescence image shows proliferating T cells (Ki67 + CD8+; yellow arrows) and the right image shows proliferating tumor cells (Ki67 + PSMA+). c Quantification of the immunofluorescence images from the tumor indicated in part (a). The top row shows changes in the number of proliferating cells per unit area (left) and changes in the percentage of proliferating cells (right). The bottom row shows percent changes in proliferating CD8+ T cells (left) and proliferating PSMA+ tumor cells (right). *P-value less than 0.05
Fig. 5
Fig. 5
a Thyroid SUVmean at baseline and after three months for all patients. Patients that experienced a thyroid related-adverse event of Gr2 or greater are shown in various colors to distinguish them from patients that did not experience a thyroid-related adverse event (black). b Receiver operating characteristic curve showing the value of thyroid SUVmean at baseline for predicting which patients will go on to experience a thyroid-related adverse event

References

    1. Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebbé C, Maio M, Binder M, Bohnsack O, Nichol G, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15:7412–7420. doi: 10.1158/1078-0432.CCR-09-1624.
    1. Yuan J, Hegde PS, Clynes R, Foukas PG, Harari A, Kleen TO. Kvistborg P, Maccalli C, Maecker HT, Page DB, et al. Novel technologies and emerging biomarkers for personalized cancer immunotherapy. J Immunother Cancer. 2016;4:3. doi: 10.1186/s40425-016-0107-3.
    1. Gainor JF, Longo DL, Chabner BA. Pharmacodynamic biomarkers: falling short of the mark? Clin Cancer Res. 2014;20:2587–2594. doi: 10.1158/1078-0432.CCR-13-3132.
    1. Gnjatic S, Bronte V, Brunet LR, Butler MO, Disis ML, Galon J, Hakansson LG, Hanks BA, Karanikas V, Khleif SN, et al. Identifying baseline immune-related biomarkers to predict clinical outcome of immunotherapy. J Immunother Cancer. 2017;5:44. doi: 10.1186/s40425-017-0243-4.
    1. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–128. doi: 10.1126/science.aaa1348.
    1. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, Walsh LA, Postow MA, Wong P, Ho TS, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–2199. doi: 10.1056/NEJMoa1406498.
    1. Masucci GV, Cesano A, Hawtin R, Janetzki S, Zhang J, Kirsch I, Dobbin KK, Alvarez J, Robbins PB, Selvan SR, et al. Validation of biomarkers to predict response to immunotherapy in cancer: volume I - pre-analytical and analytical validation. J Immunother Cancer. 2016;4:76. doi: 10.1186/s40425-016-0178-1.
    1. Becker JT, Olson BM, Johnson LE, Davies JG, Dunphy EJ, McNeel DG. DNA vaccine encoding prostatic acid phosphatase (PAP) elicits long-term T-cell responses in patients with recurrent prostate cancer. J Immunother. 2010;33:639–647. doi: 10.1097/CJI.0b013e3181dda23e.
    1. McNeel DG, Becker JT, Eickhoff JC, Johnson LE, Bradley E, Pohlkamp I, Staab MJ, Liu G, Wilding G, Olson BM. Real-time immune monitoring to guide plasmid DNA vaccination schedule targeting prostatic acid phosphatase in patients with castration-resistant prostate cancer. Clin Cancer Res. 2014;20:3692–3704. doi: 10.1158/1078-0432.CCR-14-0169.
    1. Higashikawa K, Yagi K, Watanabe K, Kamino S, Ueda M, Hiromura M, Enomoto S. 64Cu-DOTA-anti-CTLA-4 mAb enabled PET visualization of CTLA-4 on the T-cell infiltrating tumor tissues. PLoS One. 2014;9:e109866. doi: 10.1371/journal.pone.0109866.
    1. Ehlerding EB, England CG, Majewski RL, Valdovinos HF, Jiang D, Liu G, McNeel DG, Nickles RJ, Cai W. ImmunoPET imaging of CTLA-4 expression in mouse models of non-small cell lung Cancer. Mol Pharm. 2017;14:1782–1789. doi: 10.1021/acs.molpharmaceut.7b00056.
    1. Hettich M, Braun F, Bartholomä MD, Schirmbeck R, Niedermann G. High-resolution PET imaging with therapeutic antibody-based PD-1/PD-L1 checkpoint tracers. Theranostics. 2016;6:1629–1640. doi: 10.7150/thno.15253.
    1. Larimer BM, 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. doi: 10.2967/jnumed.116.173930.
    1. Tavaré R, Escuin-Ordinas H, Mok S, McCracken MN, Zettlitz KA, Salazar FB, Witte ON, Ribas A, Wu AM. An effective Immuno-PET imaging method to monitor CD8-dependent responses to immunotherapy. Cancer Res. 2016;76:73–82. doi: 10.1158/0008-5472.CAN-15-1707.
    1. Muzi M, Mankoff DA, Grierson JR, Wells JM, Vesselle H, Krohn KA. Kinetic modeling of 3′-deoxy-3′-fluorothymidine in somatic tumors: mathematical studies. J Nucl Med. 2005;46:371–380.
    1. Shields AF, Grierson JR, Dohmen BM, Machulla HJ, Stayanoff JC, Lawhorn-Crews JM, Obradovich JE, Muzik O, Mangner TJ. Imaging proliferation in vivo with [F-18]FLT and positron emission tomography. Nat Med. 1998;4:1334–1336. doi: 10.1038/3337.
    1. Muzi M, Vesselle H, Grierson JR, Mankoff DA, Schmidt RA, Peterson L, Wells JM, Krohn KA. Kinetic analysis of 3′-deoxy-3′-fluorothymidine PET studies: validation studies in patients with lung cancer. J Nucl Med. 2005;46:274–282.
    1. Backes H, Ullrich R, Neumaier B, Kracht L, Wienhard K, Jacobs AH. Noninvasive quantification of 18F-FLT human brain PET for the assessment of tumour proliferation in patients with high-grade glioma. Eur J Nucl Med Mol Imaging. 2009;36:1960–1967. doi: 10.1007/s00259-009-1244-4.
    1. Brockenbrough JS, Souquet T, Morihara JK, Stern JE, Hawes SE, Rasey JS, Leblond A, Wiens LW, Feng Q, Grierson J, Vesselle H. Tumor 3′-deoxy-3′-(18)F-fluorothymidine ((18)F-FLT) uptake by PET correlates with thymidine kinase 1 expression: static and kinetic analysis of (18)F-FLT PET studies in lung tumors. J Nucl Med. 2011;52:1181–1188. doi: 10.2967/jnumed.111.089482.
    1. Kenny LM, Vigushin DM, Al-Nahhas A, Osman S, Luthra SK, Shousha S, Coombes RC, Aboagye EO. Quantification of cellular proliferation in tumor and normal tissues of patients with breast cancer by [18F]fluorothymidine-positron emission tomography imaging: evaluation of analytical methods. Cancer Res. 2005;65:10104–10112. doi: 10.1158/0008-5472.CAN-04-4297.
    1. Hoeben BA, Troost EG, Span PN, van Herpen CM, Bussink J, Oyen WJ, Kaanders JH. 18F-FLT PET during radiotherapy or chemoradiotherapy in head and neck squamous cell carcinoma is an early predictor of outcome. J Nucl Med. 2013;54:532–540. doi: 10.2967/jnumed.112.105999.
    1. Ott K, Herrmann K, Schuster T, Langer R, Becker K, Wieder HA, Wester HJ, Siewert JR, Zum Büschenfelde CM, Buck AK, et al. Molecular imaging of proliferation and glucose utilization: utility for monitoring response and prognosis after neoadjuvant therapy in locally advanced gastric cancer. Ann Surg Oncol. 2011;18:3316–3323. doi: 10.1245/s10434-011-1743-y.
    1. Sohn HJ, Yang YJ, Ryu JS, Oh SJ, Im KC, Moon DH, Lee DH, Suh C, Lee JS, Kim SW. [18F]Fluorothymidine positron emission tomography before and 7 days after gefitinib treatment predicts response in patients with advanced adenocarcinoma of the lung. Clin Cancer Res. 2008;14:7423–7429. doi: 10.1158/1078-0432.CCR-08-0312.
    1. Chen W, Delaloye S, Silverman DH, Geist C, Czernin J, Sayre J, Satyamurthy N, Pope W, Lai A, Phelps ME, Cloughesy T. Predicting treatment response of malignant gliomas to bevacizumab and irinotecan by imaging proliferation with [18F] fluorothymidine positron emission tomography: a pilot study. J Clin Oncol. 2007;25:4714–4721. doi: 10.1200/JCO.2006.10.5825.
    1. McKinley ET, Smith RA, Tanksley JP, Washington MK, Walker R, Coffey RJ, Manning HC. [18F]FLT-PET to predict pharmacodynamic and clinical response to cetuximab therapy in Ménétrier's disease. Ann Nucl Med. 2012;26:757–763. doi: 10.1007/s12149-012-0636-x.
    1. Kishino T, Hoshikawa H, Nishiyama Y, Yamamoto Y, Mori N. Usefulness of 3′-deoxy-3'-18F-fluorothymidine PET for predicting early response to chemoradiotherapy in head and neck cancer. J Nucl Med. 2012;53:1521–1527. doi: 10.2967/jnumed.111.099200.
    1. Zander T, Scheffler M, Nogova L, Kobe C, Engel-Riedel W, Hellmich M, Papachristou I, Toepelt K, Draube A, Heukamp L, et al. Early prediction of nonprogression in advanced non-small-cell lung cancer treated with erlotinib by using [(18)F]fluorodeoxyglucose and [(18)F]fluorothymidine positron emission tomography. J Clin Oncol. 2011;29:1701–1708. doi: 10.1200/JCO.2010.32.4939.
    1. Kobe C, Scheffler M, Holstein A, Zander T, Nogova L, Lammertsma AA, Boellaard R, Neumaier B, Ullrich RT, Dietlein M, et al. Predictive value of early and late residual 18F-fluorodeoxyglucose and 18F-fluorothymidine uptake using different SUV measurements in patients with non-small-cell lung cancer treated with erlotinib. Eur J Nucl Med Mol Imaging. 2012;39:1117–1127. doi: 10.1007/s00259-012-2118-8.
    1. Wieder HA, Geinitz H, Rosenberg R, Lordick F, Becker K, Stahl A, Rummeny E, Siewert JR, Schwaiger M, Stollfuss J. PET imaging with [18F]3′-deoxy-3′-fluorothymidine for prediction of response to neoadjuvant treatment in patients with rectal cancer. Eur J Nucl Med Mol Imaging. 2007;34:878–883. doi: 10.1007/s00259-006-0292-2.
    1. McKinley ET, Watchmaker JM, Chakravarthy AB, Meyerhardt JA, Engelman JA, Walker RC, Washington MK, Coffey RJ, Manning HC. [(18)F]-FLT PET to predict early response to neoadjuvant therapy in KRAS wild-type rectal cancer: a pilot study. Ann Nucl Med. 2015;29:535–542. doi: 10.1007/s12149-015-0974-6.
    1. Hoshikawa H, Yamamoto Y, Mori T, Kishino T, Fukumura T, Samukawa Y, Mori N, Nishiyama Y. Predictive value of SUV-based parameters derived from pre-treatment (18)F-FLT PET/CT for short-term outcome with head and neck cancers. Ann Nucl Med. 2014;28:1020–1026. doi: 10.1007/s12149-014-0902-1.
    1. Herrmann K, Buck AK, Schuster T, Abbrederis K, Blümel C, Santi I, Rudelius M, Wester HJ, Peschel C, Schwaiger M, et al. Week one FLT-PET response predicts complete remission to R-CHOP and survival in DLBCL. Oncotarget. 2014;5:4050–4059. doi: 10.18632/oncotarget.1990.
    1. Challapalli A, Barwick T, Pearson RA, Merchant S, Mauri F, Howell EC, Sumpter K, Maxwell RJ, Aboagye EO, Sharma R. 3′-Deoxy-3'-18F-fluorothymidine positron emission tomography as an early predictor of disease progression in patients with advanced and metastatic pancreatic cancer. Eur J Nucl Med Mol Imaging. 2015;42:831–840. doi: 10.1007/s00259-015-3000-2.
    1. Inubushi M, Saga T, Koizumi M, Takagi R, Hasegawa A, Koto M, Wakatuki M, Morikawa T, Yoshikawa K, Tanimoto K, et al. Predictive value of 3′-deoxy-3′-[18F]fluorothymidine positron emission tomography/computed tomography for outcome of carbon ion radiotherapy in patients with head and neck mucosal malignant melanoma. Ann Nucl Med. 2013;27:1–10. doi: 10.1007/s12149-012-0652-x.
    1. Bollineni VR, Kramer GM, Jansma EP, Liu Y, Oyen WJ. A systematic review on [(18)F]FLT-PET uptake as a measure of treatment response in cancer patients. Eur J Cancer. 2016;55:81–97. doi: 10.1016/j.ejca.2015.11.018.
    1. Ribas A, Benz MR, Allen-Auerbach MS, Radu C, Chmielowski B, Seja E, Williams JL, Gomez-Navarro J, McCarthy T, Czernin J. Imaging of CTLA4 blockade-induced cell replication with (18)F-FLT PET in patients with advanced melanoma treated with tremelimumab. J Nucl Med. 2010;51:340–346. doi: 10.2967/jnumed.109.070946.
    1. Aarntzen EH, Srinivas M, De Wilt JH, Jacobs JF, Lesterhuis WJ, Windhorst AD, Troost EG, Bonenkamp JJ, van Rossum MM, Blokx WA, et al. Early identification of antigen-specific immune responses in vivo by [18F]-labeled 3′-fluoro-3′-deoxy-thymidine ([18F]FLT) PET imaging. Proc Natl Acad Sci U S A. 2011;108:18396–18399. doi: 10.1073/pnas.1113045108.
    1. McNeel DG, Eickhoff JC, Wargowski E, Zahm C, Staab MJ, Straus J, Liu G. Concurrent, but not sequential, PD-1 blockade with a DNA vaccine elicits anti-tumor responses in patients with metastatic, castration-resistant prostate cancer. Oncotarget. 2018;9:25586–25596. doi: 10.18632/oncotarget.25387.
    1. Rekoske BT, Smith HA, Olson BM, Maricque BB, McNeel DG. PD-1 or PD-L1 blockade restores antitumor efficacy following SSX2 epitope-modified DNA vaccine immunization. Cancer Immunol Res. 2015;3:946–955. doi: 10.1158/2326-6066.CIR-14-0206.
    1. Zahm CD, Colluru VT, McNeel DG. Vaccination with high-affinity epitopes impairs antitumor efficacy by increasing PD-1 expression on CD8. Cancer Immunol Res. 2017;5:630–641. doi: 10.1158/2326-6066.CIR-16-0374.
    1. Scher HI, Halabi S, Tannock I, Morris M, Sternberg CN, Carducci MA, Eisenberger MA, Higano C, Bubley GJ, Dreicer R, et al. Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the prostate Cancer clinical trials working group. J Clin Oncol. 2008;26:1148–1159. doi: 10.1200/JCO.2007.12.4487.
    1. Thie JA. Understanding the standardized uptake value, its methods and implications for usage. J Nucl Med. 2004;45:1431–1434.
    1. Scarpelli M, Eickhoff J, Cuna E, Perlman S, Jeraj R. Optimal transformations leading to normal distributions of positron emission tomography standardized uptake values. Phys Med Biol. 2018;63:035021. doi: 10.1088/1361-6560/aaa175.
    1. Schröder MS, Culhane AC, Quackenbush J, Haibe-Kains B. Survcomp: an R/Bioconductor package for performance assessment and comparison of survival models. Bioinformatics. 2011;27:3206–3208. doi: 10.1093/bioinformatics/btr511.
    1. Pencina MJ, D'Agostino RB. Overall C as a measure of discrimination in survival analysis: model specific population value and confidence interval estimation. Stat Med. 2004;23:2109–2123. doi: 10.1002/sim.1802.
    1. Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez JC, Müller M. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics. 2011;12:77. doi: 10.1186/1471-2105-12-77.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, 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. doi: 10.1016/j.ejca.2008.10.026.
    1. Osorio JC, Ni A, Chaft JE, Pollina R, Kasler MK, Stephens D, Rodriguez C, Cambridge L, Rizvi H, Wolchok JD, et al. Antibody-mediated thyroid dysfunction during T-cell checkpoint blockade in patients with non-small-cell lung cancer. Ann Oncol. 2017;28:583–589.
    1. Michot JM, Bigenwald C, Champiat S, Collins M, Carbonnel F, Postel-Vinay S, Berdelou A, Varga A, Bahleda R, Hollebecque A, et al. Immune-related adverse events with immune checkpoint blockade: a comprehensive review. Eur J Cancer. 2016;54:139–148. doi: 10.1016/j.ejca.2015.11.016.
    1. Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 2018;378:158–168. doi: 10.1056/NEJMra1703481.

Source: PubMed

3
Subskrybuj