F18-FDG PET/CT imaging early predicts pathologic complete response to induction chemoimmunotherapy of locally advanced head and neck cancer: preliminary single-center analysis of the checkrad-cd8 trial

M Beck, J Hartwich, M Eckstein, D Schmidt, A O Gostian, S Müller, S Rutzner, U S Gaipl, J von der Grün, T Illmer, M G Hautmann, G Klautke, J Döscher, T Brunner, B Tamaskovics, A Hartmann, H Iro, T Kuwert, R Fietkau, M Hecht, S Semrau, M Beck, J Hartwich, M Eckstein, D Schmidt, A O Gostian, S Müller, S Rutzner, U S Gaipl, J von der Grün, T Illmer, M G Hautmann, G Klautke, J Döscher, T Brunner, B Tamaskovics, A Hartmann, H Iro, T Kuwert, R Fietkau, M Hecht, S Semrau

Abstract

Aim: In the CheckRad-CD8 trial patients with locally advanced head and neck squamous cell cancer are treated with a single cycle of induction chemo-immunotherapy (ICIT). Patients with pathological complete response (pCR) in the re-biopsy enter radioimmunotherapy. Our goal was to study the value of F-18-FDG PET/CT in the prediction of pCR after induction therapy.

Methods: Patients treated within the CheckRad-CD8 trial that additionally received FDG- PET/CT imaging at the following two time points were included: 3-14 days before (pre-ICIT) and 21-28 days after (post-ICIT) receiving ICIT. Tracer uptake in primary tumors (PT) and suspicious cervical lymph nodes (LN +) was measured using different quantitative parameters on EANM Research Ltd (EARL) accredited PET reconstructions. In addition, mean FDG uptake levels in lymphatic and hematopoietic organs were examined. Percent decrease (Δ) in FDG uptake was calculated for all parameters. Biopsy of the PT post-ICIT acquired after FDG-PET/CT served as reference. The cohort was divided in patients with pCR and residual tumor (ReTu).

Results: Thirty-one patients were included. In ROC analysis, ΔSUVmax PT performed best (AUC = 0.89) in predicting pCR (n = 17), with a decline of at least 60% (sensitivity, 0.77; specificity, 0.93). Residual SUVmax PT post-ICIT performed best in predicting ReTu (n = 14), at a cutpoint of 6.0 (AUC = 0.91; sensitivity, 0.86; specificity, 0.88). Combining two quantitative parameters (ΔSUVmax ≥ 50% and SUVmax PT post-ICIT ≤ 6.0) conferred a sensitivity of 0.81 and a specificity of 0.93 for determining pCR. Background activity in lymphatic organs or uptake in suspected cervical lymph node metastases lacked significant predictive value.

Conclusion: FDG-PET/CT can identify patients with pCR after ICIT via residual FDG uptake levels in primary tumors and the related changes compared to baseline. FDG-uptake in LN + had no predictive value.

Trial registry: ClinicalTrials.gov identifier: NCT03426657.

Keywords: FDG-PET/CT; HNSCC; Head neck cancer; Immunotherapy; Induction therapy.

Conflict of interest statement

M.B. conflict of interest with Novartis (travel expenses); Bayer (advisory role, honoraria); SIRTEX (travel expenses). J.H. has no conflict of interest. M.E. conflict of interest with Diaceutics (employment, honoraria, advisory role, speakers’ bureau, travel expenses); AstraZeneca (honoraria, advisory role, speakers’ bureau, travel expenses); Roche (honoraria, travel expenses); MSD (honoraria, speakers’ bureau); GenomicHealth (honoraria, advisory role, speakers bureau, travel expenses); Astellas (honoraria, speakers’ bureau); Janssen-Cilag (honoraria, advisory role, research funding, travel expenses); Stratifyer (research funding, patents). D.S. has no conflict of interest. A.O.G. has no conflict of interest. S.M. has no conflict of interest. S.R. conflict of interest with AstraZeneca (research funding); MSD (research funding). U.S.G. conflict of interest with AstraZeneca (advisory role, research funding); BMS (advisory role); MSD (research funding); Sennewald Medizintechnik (travel expenses). J.G. has no conflict of interest. I.T. has no conflict of interest. M.G.H. conflict of interest with Roche (stock, honoraria); Varian (stock); Sanofi (honoraria); AstraZeneca (honoraria, travel expenses); BMS (honoraria, advisory role); MSD (honoraria, advisory role); Merck Serono (honoraria); Celgene (honoraria). G.K. conflict of interest with BMS (advisory role); Lilly (advisory role); Roche (advisory role). J.D. has no conflict of interest. B.T. conflict of interest with BMS (honoraria, advisory role, travel expenses); Merck Serono (honoraria, advisory role); MSD (travel expenses). A.H. conflict of interest with BMS (honoraria, advisory role); MSD (honoraria, advisory role); Roche (honoraria, advisory role, research funding); AstraZeneca (honoraria, advisory role, research funding); Boehringer Ingelheim (honoraria); Abbvie (honoraria); Cepheid (advisory role, research funding); Quiagen (advisory role); Janssen-Cilag (honoraria, advisory role, research funding); Ipsen (honoraria, advisory role); NanoString Technologies (advisory role, research funding, expert testimony); Illumina (advisory role); 3DHistech (advisory role); Diaceutics (advisory role); BioNTech (research funding). H.I. has no conflict of interest. T.K. conflict of interest with Siemens Healthineers (advisory role, honoraria, speakers’ bureau, travel expenses, research funding). R.F. conflict of interest with MSD (honoraria, advisory role, research funding, travel expenses); Fresenius (honoraria); BrainLab (honoraria); AstraZeneca (honoraria, advisory role, research funding, travel expenses); Merck Serono (advisory role, research funding, travel expenses); Novocure (advisory role, speakers’ bureau, research funding); Sennewald (speakers’ bureau, travel expenses). The other authors declare no conflicts of interest. M.H. conflict of interest with Merck Serono (advisory role, speakers’ bureau, honoraria, travel expenses, research funding); MSD (advisory role, speakers’ bureau, travel expenses, research funding); AstraZeneca (research funding); Novartis (research funding); BMS (advisory role, honoraria, speakers’ bureau); Teva (travel expenses). S.S. conflict of interest with Strycker (stock); Varian (stock); Abbot (stock); Crispr Techn. (stock); Pfitzer (stock); Merck Serono (stock); Symrise (stock); Ortho (honoraria, advisory role, speakers’ bureau, research funding, travel expenses); PharmaMar (speakers’ bureau, travel expenses); Haema (speakers’ bureau).

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Flowchart of the CheckRad CD-8 trial design BSA body surface area, pCR pathological complete remission; ReTu residual tumor
Fig. 2
Fig. 2
Upper row: Representative patient with (A) cT2 cN2c HNSCC of hypopharynx prior to induction chemoimmunotherapy. FDG-PET/CT records a high baseline metabolic activity high (SUVmax, 19.8). Histology shows a moderately to poorly differentiated non-keratinizing squamous cell carcinoma (B) HE staining with a low levels of stromal and intratumoral infiltration with cytotoxic T-Cells (CD8 immunohistochemistry) (C). Lower row: Same patient after ICIT: (D) marked decline in uptake by primary tumor (SUVmax, 2.8; ΔSUVpeak, 85.9%). Histology shows a densely packed lymphoid stroma in the HE stain (E) with CD8 positive lymphocytes after immunolabeling (D) but without evidence of viable tumor cells (F). PT primary tumor, SUV standardized uptake value, HE stain hematoxylin and eosin stain
Fig. 3
Fig. 3
Receiver operating characteristic (ROC) curve plotted for complete remission in subset of patients with nodal metastasis (n = 25)

References

    1. Fietkau R, Hecht M, Hofner B, Lubgan D, Iro H, Gefeller O, et al. Randomized phase-III-trial of concurrent chemoradiation for locally advanced head and neck cancer comparing dose reduced radiotherapy with paclitaxel/cisplatin to standard radiotherapy with fluorouracil/cisplatin: the PacCis-trial. Radiother Oncol. 2020;144:209–217. doi: 10.1016/j.radonc.2020.01.016.
    1. Ang KK, Zhang Q, Rosenthal DI, Nguyen-Tan PF, Sherman EJ, Weber RS, et al. Randomized phase III trial of concurrent accelerated radiation plus cisplatin with or without cetuximab for stage III to IV head and neck carcinoma: RTOG 0522. J Clin Oncol. 2014;32(27):2940–2950. doi: 10.1200/JCO.2013.53.5633.
    1. Mogadas S, Busch CJ, Pflug C, Hanken H, Krull A, Petersen C, et al. Influence of radiation dose to pharyngeal constrictor muscles on late dysphagia and quality of life in patients with locally advanced oropharyngeal carcinoma. Strahlenther Onkol. 2020;196(6):522–529. doi: 10.1007/s00066-019-01572-0.
    1. Forastiere AA, Zhang Q, Weber RS, Maor MH, Goepfert H, Pajak TF, et al. Long-term results of RTOG 91–11: a comparison of three nonsurgical treatment strategies to preserve the larynx in patients with locally advanced larynx cancer. J Clin Oncol. 2013;31(7):845–852. doi: 10.1200/JCO.2012.43.6097.
    1. Breheret M, Lubgan D, Haderlein M, Hecht M, Traxdorf M, Schmidt D, et al. Single-cycle induction chemotherapy before chemoradiotherapy or surgery in functionally inoperable head and neck squamous cell carcinoma: 10-year results. Eur Arch Otorhinolaryngol. 2020;277(1):245–254. doi: 10.1007/s00405-019-05665-5.
    1. Semrau S, Haderlein M, Schmidt D, Lell M, Wolf W, Waldfahrer F, et al. Single-cycle induction chemotherapy followed by chemoradiotherapy or surgery in patients with head and neck cancer: what are the best predictors of remission and prognosis? Cancer. 2015;121(8):1214–1222. doi: 10.1002/cncr.29188.
    1. Semrau S, Schmidt D, Lell M, Waldfahrer F, Lettmaier S, Kuwert T, et al. Results of chemoselection with short induction chemotherapy followed by chemoradiation or surgery in the treatment of functionally inoperable carcinomas of the pharynx and larynx. Oral Oncol. 2013;49(5):454–460. doi: 10.1016/j.oraloncology.2012.12.008.
    1. Tanadini-Lang S, Balermpas P, Guckenberger M, Pavic M, Riesterer O, Vuong D, et al. Radiomic biomarkers for head and neck squamous cell carcinoma. Strahlenther Onkol. 2020;196(10):868–878. doi: 10.1007/s00066-020-01638-4.
    1. Budach W, Bolke E, Kammers K, Gerber PA, Orth K, Gripp S, et al. Induction chemotherapy followed by concurrent radio-chemotherapy versus concurrent radio-chemotherapy alone as treatment of locally advanced squamous cell carcinoma of the head and neck (HNSCC): a meta-analysis of randomized trials. Radiother Oncol. 2016;118(2):238–243. doi: 10.1016/j.radonc.2015.10.014.
    1. Semrau S, Schmidt D, Hecht M, Haderlein M, Kitzsteiner C, Muller S, et al. Classification of three prognostically different groups of head and neck cancer patients based on their metabolic response to induction chemotherapy (IC-1) Oral Oncol. 2020;100:104479. doi: 10.1016/j.oraloncology.2019.104479.
    1. Kikuchi M, Nakamoto Y, Shinohara S, Fujiwara K, Yamazaki H, Kanazawa Y, et al. Early evaluation of neoadjuvant chemotherapy response using FDG-PET/CT predicts survival prognosis in patients with head and neck squamous cell carcinoma. Int J Clin Oncol. 2013;18(3):402–410. doi: 10.1007/s10147-012-0393-9.
    1. Franklin C, Livingstone E, Roesch A, Schilling B, Schadendorf D. Immunotherapy in melanoma: recent advances and future directions. Eur J Surg Oncol. 2017;43(3):604–611. doi: 10.1016/j.ejso.2016.07.145.
    1. Considine B, Hurwitz ME. Current status and future directions of immunotherapy in renal cell carcinoma. Curr Oncol Rep. 2019;21(4):34. doi: 10.1007/s11912-019-0779-1.
    1. Burtness B, Harrington KJ, Greil R, Soulieres D, Tahara M, de Castro G, et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study. Lancet. 2019;394(10212):1915–28. doi: 10.1016/S0140-6736(19)32591-7.
    1. Ott PA, Hodi FS, Robert C. CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin Cancer Res: Off J Am Assoc Cancer Res. 2013;19(19):5300–5309. doi: 10.1158/1078-0432.CCR-13-0143.
    1. Bleeker-Rovers CP, de Kleijn EM, Corstens FH, van der Meer JW, Oyen WJ. Clinical value of FDG PET in patients with fever of unknown origin and patients suspected of focal infection or inflammation. Eur J Nucl Med Mol Imaging. 2004;31(1):29–37. doi: 10.1007/s00259-003-1338-3.
    1. Cho SY, Huff DT, Jeraj R, Albertini MR. FDG PET/CT for assessment of immune therapy: opportunities and understanding pitfalls. Semin Nucl Med. 2020;50(6):518–531. doi: 10.1053/j.semnuclmed.2020.06.001.
    1. Tsai KK, Pampaloni MH, Hope C, Algazi AP, Ljung BM, Pincus L, et al. Increased FDG avidity in lymphoid tissue associated with response to combined immune checkpoint blockade. J Immunother Cancer. 2016;4:58. doi: 10.1186/s40425-016-0162-9.
    1. Hecht M, Gostian AO, Eckstein M, Rutzner S, von der Grun J, Illmer T, et al. Safety and efficacy of single cycle induction treatment with cisplatin/docetaxel/ durvalumab/tremelimumab in locally advanced HNSCC: first results of checkrad-CD8. J Immunother Cancer. 2020;8(2):e001378. doi: 10.1136/jitc-2020-001378.
    1. Hecht M, Eckstein M, Rutzner S, von der Grun J, Illmer T, Klautke G, et al. Induction chemoimmunotherapy followed by CD8+ immune cell-based patient selection for chemotherapy-free radioimmunotherapy in locally advanced head and neck cancer. J Immunother Cancer. 2022;10(1):e003747. doi: 10.1136/jitc-2021-003747.
    1. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, et al. New guidelines to evaluate the response to treatment in solid tumors European organization for research and treatment of cancer, national cancer institute of the United States, national cancer institute of canada. J National Cancer Inst. 2000;92(3):205–16. doi: 10.1093/jnci/92.3.205.
    1. Wahl RL, Jacene H, Kasamon Y, Lodge MA. From RECIST to PERCIST: evolving considerations for PET response criteria in solid tumors. J Nuclear Med: Off Publ, Soc Nuclear Med. 2009;50(Suppl 1):122S–S150. doi: 10.2967/jnumed.108.057307.
    1. Gilles R, de Geus-Oei LF, Mulders PF, Oyen WJ. Immunotherapy response evaluation with (18)F-FDG-PET in patients with advanced stage renal cell carcinoma. World J Urol. 2013;31(4):841–846. doi: 10.1007/s00345-011-0723-y.
    1. Aide N, Hicks RJ, Le Tourneau C, Lheureux S, Fanti S, Lopci E. 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(1):238–250. doi: 10.1007/s00259-018-4171-4.
    1. Carter BW, Bhosale PR, Yang WT. Immunotherapy and the role of imaging. Cancer. 2018;124(14):2906–2922. doi: 10.1002/cncr.31349.
    1. Shields AF, Jacobs PM, Sznol M, Graham MM, Germain RN, Lum LG, et al. Immune modulation therapy and imaging: workshop report. J Nuclear Med: Off Publ, Soc Nuclear Med. 2018;59(3):410–417. doi: 10.2967/jnumed.117.195610.
    1. Zhou JG, Donaubauer AJ, Frey B, Becker I, Rutzner S, Eckstein M, et al. Prospective development and validation of a liquid immune profile-based signature (LIPS) to predict response of patients with recurrent/metastatic cancer to immune checkpoint inhibitors. J Immunother Cancer. 2021;9(2):e001845. doi: 10.1136/jitc-2020-001845.
    1. Humbert O, Cadour N, Paquet M, Schiappa R, Poudenx M, Chardin D, et al. (18)FDG PET/CT in the early assessment of non-small cell lung cancer response to immunotherapy: frequency and clinical significance of atypical evolutive patterns. Eur J Nucl Med Mol Imaging. 2020;47(5):1158–1167. doi: 10.1007/s00259-019-04573-4.
    1. 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(1):8. doi: 10.1186/s13550-019-0473-1.

Source: PubMed

Подписаться