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

Michael D Farwell, Raymond F Gamache, Hasan Babazada, Matthew D Hellmann, James J Harding, Ron Korn, Alessandro Mascioni, William Le, Ian Wilson, Michael S Gordon, Anna M Wu, Gary A Ulaner, Jedd D Wolchok, Michael A Postow, Neeta Pandit-Taskar, Michael D Farwell, Raymond F Gamache, Hasan Babazada, Matthew D Hellmann, James J Harding, Ron Korn, Alessandro Mascioni, William Le, Ian Wilson, Michael S Gordon, Anna M Wu, Gary A Ulaner, Jedd D Wolchok, Michael A Postow, Neeta Pandit-Taskar

Abstract

There is a need for in vivo diagnostic imaging probes that can noninvasively measure tumor-infiltrating CD8+ leukocytes. Such imaging probes could be used to predict early response to cancer immunotherapy, help select effective single or combination immunotherapies, and facilitate the development of new immunotherapies or immunotherapy combinations. This study was designed to optimize conditions for performing CD8 PET imaging with 89Zr-Df-IAB22M2C and determine whether CD8 PET imaging could provide a safe and effective noninvasive method of visualizing the whole-body biodistribution of CD8+ leukocytes. Methods: We conducted a phase 1 first-in-humans PET imaging study using an anti-CD8 radiolabeled minibody, 89Zr-Df-IAB22M2C, to detect whole-body and tumor CD8+ leukocyte distribution in patients with metastatic solid tumors. Patients received 111 MBq of 89Zr-Df-IAB22M2C followed by serial PET scanning over 5-7 d. A 2-stage design included a dose-escalation phase and a dose-expansion phase. Biodistribution, radiation dosimetry, and semiquantitative evaluation of 89Zr-Df-IAB22M2C uptake were performed in all patients. Results: Fifteen subjects with metastatic melanoma, non-small cell lung cancer, and hepatocellular carcinoma were enrolled. No drug-related adverse events or abnormal laboratory results were noted except for a transient increase in antidrug antibodies in 1 subject. 89Zr-Df-IAB22M2C accumulated in tumors and CD8-rich tissues (e.g., spleen, bone marrow, nodes), with maximum uptake at 24-48 h after injection and low background activity in CD8-poor tissues (e.g., muscle and lung). Radiotracer uptake in tumors was noted in 10 of 15 subjects, including 7 of 8 subjects on immunotherapy, 1 of 2 subjects on targeted therapy, and 2 of 5 treatment-naïve subjects. In 3 patients with advanced melanoma or hepatocellular carcinoma on immunotherapy, posttreatment CD8 PET/CT scans demonstrated increased 89Zr-Df-IAB22M2C uptake in tumor lesions, which correlated with response. Conclusion: CD8 PET imaging with 89Zr-Df-IAB22M2C is safe and has the potential to visualize the whole-body biodistribution of CD8+ leukocytes in tumors and reference tissues, and may predict early response to immunotherapy.

Trial registration: ClinicalTrials.gov NCT03107663.

Keywords: 89Zr-Df-IAB22M2C; CD8+ T cell; PET imaging; immunotherapy; minibody.

© 2022 by the Society of Nuclear Medicine and Molecular Imaging.

Figures

Graphical abstract
Graphical abstract
FIGURE 1.
FIGURE 1.
Serum clearance and biodistribution of 89Zr-Df-IAB22M2C. (A) Serum clearance of 89Zr-Df-IAB22M2C based on enzyme-linked immunosorbent assay measurements (limit of detection = 5 ng/mL). No minibody was detected in serum at the 0.2-mg dose. (B) Whole-body PET images of a patient at various times after injection of 89Zr-Df-IAB22M2C (1.5-mg minibody dose) demonstrating the distribution of 89Zr-Df-IAB22M2C in normal tissues and uptake in a nodal metastasis in the right neck (arrow), with good visualization of uptake in the nodal metastasis at 24–48 h after injection.
FIGURE 2.
FIGURE 2.
89Zr-Df-IAB22M2C uptake in normal tissues and tumor lesions versus time. (A) 89Zr-Df-IAB22M2C uptake in CD8-rich reference tissues in patients administered 0.5 and 1.5 mg of minibody mass. (B) 89Zr-Df-IAB22M2C uptake in CD8-poor reference tissues in patients administered 0.5 and 1.5 mg of minibody mass. (C) Box and whisker plots of 89Zr-Df-IAB22M2C uptake in tumor lesions from all subjects (n = 15). Boxes outline first and third quartile values. Median SUVMAX values are indicated by horizontal line and mean SUVMAX values are indicated with +. Outlier values are indicated by dots. (D) 89Zr-Df-IAB22M2C mean tumor uptake in patients who received 0.5 and 1.5 mg of minibody mass. BM = bone marrow; LN = lymph nodes.
FIGURE 3.
FIGURE 3.
A 77-y-old man with metastatic melanoma treated with pembrolizumab. CT and fused 18F-FDG PET/CT images (left) acquired at approximately 8 mo after initiation of immunotherapy demonstrate 2 18F-FDG–avid nodal metastases in right neck (SUVMAX = 8.0, top image; SUVMAX = 16.8, bottom image), which could represent viable metastases. Corresponding CT and fused CD8 PET/CT images (right) obtained at 1 mo after 18F-FDG PET/CT demonstrate significant tracer activity in both metastases (SUVMAX = 5.4, top image; SUVMAX = 14.6, bottom image), which suggests that some of the 18F-FDG activity could be due to tumor-infiltrating CD8+ T cells rather than tumor cells. Follow-up imaging over the next 6 mo demonstrated stable disease, supportive of this hypothesis.
FIGURE 4.
FIGURE 4.
A 71-y-old man with locally advanced stage III melanoma treated with pembrolizumab. Baseline CT and fused 18F-FDG PET/CT images (left) demonstrate 2 18F-FDG–avid metastases in left axilla (SUVMAX = 10.0, medial node; SUVMAX = 7.6, lateral node). CT and fused CD8 PET/CT images (middle) obtained at 28 d after start of immunotherapy demonstrate increased tracer activity in both metastases (SUVMAX = 9.5, medial node; SUVMAX = 10.0, lateral node), suggestive of tumor infiltration by CD8+ T cells. Follow-up imaging with contrast-enhanced CT (right) demonstrated complete response to therapy.

References

    1. Haslam A, Prasad V. Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw Open. 2019;2:e192535.
    1. Brahmer JR, Tykodi SS, Chow LQ, et al. . Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–2465.
    1. Topalian SL, Hodi FS, Brahmer JR, et al. . Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–2454.
    1. Azimi F, Scolyer RA, Rumcheva P, et al. . Tumor-infiltrating lymphocyte grade is an independent predictor of sentinel lymph node status and survival in patients with cutaneous melanoma. J Clin Oncol. 2012;30:2678–2683.
    1. Gooden MJ, de Bock GH, Leffers N, Daemen T, Nijman HW. The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Br J Cancer. 2011;105:93–103.
    1. Huang AC, Orlowski RJ, Xu X, et al. . A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat Med. 2019;25:454–461.
    1. Ribas A, Dummer R, Puzanov I, et al. . Oncolytic virotherapy promotes intratumoral T cell infiltration and improves anti-PD-1 immunotherapy. Cell. 2018;174:1031–1032.
    1. Tumeh PC, Harview CL, Yearley JH, et al. . PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571.
    1. Griessinger CM, Olafsen T, Mascioni A, et al. . The PET-tracer (89)Zr-Df-IAB22M2C enables monitoring of intratumoral CD8 T-cell infiltrates in tumor-bearing humanized mice after T-cell bispecific antibody treatment. Cancer Res. 2020;80:2903–2913.
    1. Olafsen T, Torgov M, Zhang GG, et al. . PET imaging of cytotoxic human T cells using an 89Zr-labeled anti-CD8 minibody. J Immunother Cancer. 2015;3(suppl 2):P388.
    1. Olafsen T, Jiang ZK, Romero J, et al. . Sensitivity of 89Zr-labeled anti-CD8 minibody for PET imaging of infiltrating CD8+ T cells [abstract]. Cancer Res. 2016;76:LB-188.
    1. Pandit-Taskar N, Postow MA, Hellmann MD, et al. . First-in-humans imaging with 89Zr-Df-IAB22M2C anti-CD8 minibody in patients with solid malignancies: preliminary pharmacokinetics, biodistribution, and lesion targeting. J Nucl Med. 2020;61:512–519.
    1. Holland JP, Caldas-Lopes E, Divilov V, et al. . Measuring the pharmacodynamic effects of a novel Hsp90 inhibitor on HER2/neu expression in mice using Zr-DFO-trastuzumab. PLoS One. 2010;5:e8859.
    1. Holland JP, Divilov V, Bander NH, Smith-Jones PM, Larson SM, Lewis JS. 89Zr-DFO-J591 for immunoPET of prostate-specific membrane antigen expression in vivo. J Nucl Med. 2010;51:1293–1300.
    1. Holland JP, Sheh Y, Lewis JS. Standardized methods for the production of high specific-activity zirconium-89. Nucl Med Biol. 2009;36:729–739.
    1. Stabin MG, Sparks RB, Crowe E. OLINDA/EXM: the second-generation personal computer software for internal dose assessment in nuclear medicine. J Nucl Med. 2005;46:1023–1027.
    1. Siegel JA, Thomas SR, Stubbs JB, et al. . MIRD pamphlet no. 16: techniques for quantitative radiopharmaceutical biodistribution data acquisition and analysis for use in human radiation dose estimates. J Nucl Med. 1999;40:37S–61S.
    1. International Commission on Radiological Protection (ICRP). The 2007 Recommendations of the International Commission on Radiological Protection. ICRP publication 103. Ann ICRP. 2007;37:1–332.
    1. Börjesson PK, Jauw YW, de Bree R, et al. . Radiation dosimetry of 89Zr-labeled chimeric monoclonal antibody U36 as used for immuno-PET in head and neck cancer patients. J Nucl Med. 2009;50:1828–1836.
    1. Laforest R, Lapi SE, Oyama R, et al. . [89Zr]Trastuzumab: evaluation of radiation dosimetry, safety, and optimal imaging parameters in women with HER2-positive breast cancer. Mol Imaging Biol. 2016;18:952–959.
    1. Lindenberg L, Adler S, Turkbey IB, et al. . Dosimetry and first human experience with 89Zr-panitumumab. Am J Nucl Med Mol Imaging. 2017;7:195–203.
    1. Pandit-Taskar N, O’Donoghue JA, Ruan S, et al. . First-in-human imaging with 89Zr-Df-IAB2M anti-PSMA minibody in patients with metastatic prostate cancer: pharmacokinetics, biodistribution, dosimetry, and lesion uptake. J Nucl Med. 2016;57:1858–1864.
    1. Ulaner GA, Lyashchenko SK, Riedl C, et al. . First-in-human human epidermal growth factor receptor 2-targeted imaging using 89Zr-pertuzumab PET/CT: dosimetry and clinical application in patients with breast cancer. J Nucl Med. 2018;59:900–906.
    1. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541:321–330.
    1. Cho SY, Lipson EJ, Im HJ, 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;58:1421–1428.
    1. Iravani A, Osman MM, Weppler AM, et al. . FDG PET/CT for tumoral and systemic immune response monitoring of advanced melanoma during first-line combination ipilimumab and nivolumab treatment. Eur J Nucl Med Mol Imaging. 2020;47:2776–2786.
    1. Ito K, Teng R, Schoder H, et al. . 18F-FDG PET/CT for monitoring of ipilimumab therapy in patients with metastatic melanoma. J Nucl Med. 2019;60:335–341.
    1. Nobashi T, Baratto L, Reddy SA, et al. . Predicting response to immunotherapy by evaluating tumors, lymphoid cell-rich organs, and immune-related adverse events using FDG-PET/CT. Clin Nucl Med. 2019;44:e272–e279.
    1. Scarpelli M, Zahm C, Perlman S, McNeel DG, Jeraj R, Liu G. FLT PET/CT imaging of metastatic prostate cancer patients treated with pTVG-HP DNA vaccine and pembrolizumab. J Immunother Cancer. 2019;7:23.
    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.
    1. Umeda Y, Morikawa M, Anzai M, et al. . Predictive value of integrated 18F-FDG PET/MRI in the early response to nivolumab in patients with previously treated non-small cell lung cancer. J Immunother Cancer. 2020;8: e000349.
    1. Yeh R, Trager MH, Rizk EM, et al. . FLT-PET at 6 weeks predicts response assessed by CT at 12 weeks in melanoma patients treated with pembrolizumab. Clin Nucl Med. 2020;45:267–275.
    1. de Ruijter LK, van de Donk PP, Hooiveld-Noeken JS, et al. . 89ZED88082A PET imaging to visualize CD8+ T cells in patients with cancer treated with immune checkpoint inhibitor [abstract]. Cancer Res. 2021;81:LB037.

Source: PubMed

3
Abonner