Skip to main content

Advertisement

Log in

A phase I study of a PARP1-targeted topical fluorophore for the detection of oral cancer

  • Original Article
  • Published:
European Journal of Nuclear Medicine and Molecular Imaging Aims and scope Submit manuscript

Abstract

Background

Visual inspection and biopsy is the current standard of care for oral cancer diagnosis, but is subject to misinterpretation and consequently to misdiagnosis. Topically applied PARPi-FL is a molecularly specific, fluorescent contrast-based approach that may fulfill the unmet need for a simple, in vivo, non-invasive, cost-effective, point-of-care method for the early diagnosis of oral cancer. Here, we present results from a phase I safety and feasibility study on fluorescent, topically applied PARPi-FL. Twelve patients with a histologically proven oral squamous cell carcinoma (OSCC) gargled a PARPi-FL solution for 60 s (15 mL, 100 nM, 250 nM, 500 nM, or 1000 nM), followed by gargling a clearing solution for 60 s. Fluorescence measurements of the lesion and surrounding oral mucosa were taken before PARPi-FL application, after PARPi-FL application, and after clearing. Blood pressure, oxygen levels, clinical chemistry, and CBC were obtained before and after tracer administration.

Results

PARPi-FL was well-tolerated by all patients without any safety concerns. When analyzing the fluorescence signal, all malignant lesions showed a significant differential in contrast after administration of PARPi-FL, with the highest increase occurring at the highest dose level (1000 nM), where all patients had a tumor-to-margin fluorescence signal ratio of >3. A clearing step was essential to increase signal specificity, as it clears unbound PARPi-FL trapped in normal anatomical structures. PARPi-FL tumor cell specificity was confirmed by ex vivo tabletop confocal microscopy. We have demonstrated that the fluorescence signal arose from the nuclei of tumor cells, endorsing our macroscopic findings.

Conclusions

A PARPi-FL swish & spit solution is a rapid and non-invasive diagnostic tool that preferentially localizes fluorescent contrast to OSCC. This technique holds promise for the early detection of OSCC based on in vivo optical evaluation and targeted biopsy of suspicious lesions in the oral cavity.

Trial registration

Clinicaltrials.gov—NCT03085147, registered on March 21st, 2017.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Abbreviations

PARP:

Poly(ADP-ribose)polymerase

OSCC:

Squamous cell carcinoma of the oral cavity

IHC:

Immunohistochemistry

PEG 300:

Polyethylene glycol 300

ROI:

Regions of interest

TMR:

Tumor to normal mucosal ratios

DAB:

3,3′-Diaminobenzidin

LMR:

Lesion to margin ratio

LED:

Light-emitting diode

MSK:

Memorial Sloan Kettering

References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70(1):7–30.

    Article  Google Scholar 

  2. Chakraborty D, Natarajan C, Mukherjee A. Advances in oral cancer detection. Adv Clin Chem. 2019;91:181–200.

    Article  CAS  Google Scholar 

  3. Bosetti C, Carioli G, Santucci C, Bertuccio P, Gallus S, Garavello W, et al. Global trends in oral and pharyngeal cancer incidence and mortality. Int J Cancer. 2020;147(4):1040–9.

    Article  CAS  Google Scholar 

  4. Noonan B. Understanding the reasons why patients delay seeking treatment for oral cancer symptoms from a primary health care professional: an integrative literature review. Eur J Oncol Nurs. 2014;18(1):118–24.

    Article  Google Scholar 

  5. LeHew CW, Epstein JB, Kaste LM, Choi YK. Assessing oral cancer early detection: clarifying dentists’ practices. J Public Health Dent. 2010;70(2):93–100.

    PubMed  Google Scholar 

  6. Lingen MW, Tampi MP, Urquhart O, Abt E, Agrawal N, Chaturvedi AK, et al. Adjuncts for the evaluation of potentially malignant disorders in the oral cavity: diagnostic test accuracy systematic review and meta-analysis-a report of the American Dental Association. J Am Dent Assoc. 2017;148(11):797–813.e52.

    Article  Google Scholar 

  7. Montero PH, Patel SG. Cancer of the oral cavity. Surg Oncol Clin N Am. 2015;24(3):491–508.

    Article  Google Scholar 

  8. Kaur J, Matta A, Kak I, Srivastava G, Assi J, Leong I, et al. S100A7 overexpression is a predictive marker for high risk of malignant transformation in oral dysplasia. Int J Cancer. 2014;134(6):1379–88.

    Article  CAS  Google Scholar 

  9. Strome A, Kossatz S, Zanoni DK, Rajadhyaksha M, Patel S, Reiner T. Current practice and emerging molecular imaging technologies in oral cancer screening. Mol Imaging. 2018;17:1536012118808644.

    Article  Google Scholar 

  10. Kaur J, Jacobs R, Huang Y, Salvo N, Politis C. Salivary biomarkers for oral cancer and pre-cancer screening: a review. Clin Oral Investig. 2018;22(2):633–40.

    Article  CAS  Google Scholar 

  11. Fedele S. Diagnostic aids in the screening of oral cancer. Head neck Oncol. 2009;1:5.

    Article  Google Scholar 

  12. Catarino R, Schafer S, Vassilakos P, Petignat P, Arbyn M. Accuracy of combinations of visual inspection using acetic acid or lugol iodine to detect cervical precancer: a meta-analysis. BJOG. 2018;125(5):545–53.

    Article  CAS  Google Scholar 

  13. Sridharan G, Shankar AA. Toluidine blue: a review of its chemistry and clinical utility. J Oral Maxillofac Pathol. 2012;16(2):251–5.

    Article  Google Scholar 

  14. Peterson G, Zanoni DK, Ardigo M, Migliacci JC, Patel SG, Rajadhyaksha M. Feasibility of a video-mosaicking approach to extend the field-of-view for reflectance confocal microscopy in the oral cavity in vivo. Lasers Surg Med. 2019. https://doi.org/10.1002/lsm.23090.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Pavlova I, Williams M, El-Naggar A, Richards-Kortum R, Gillenwater A. Understanding the biological basis of autofluorescence imaging for oral cancer detection: high-resolution fluorescence microscopy in viable tissue. Clin Cancer Res. 2008;14(8):2396–404.

    Article  Google Scholar 

  16. Reiner T, Lacy J, Keliher EJ, Yang KS, Ullal A, Kohler RH, et al. Imaging therapeutic PARP inhibition in vivo through bioorthogonally developed companion imaging agents. Neoplasia (New York, NY). 2012;14(3):169–77.

    Article  CAS  Google Scholar 

  17. Dubach JM, Kim E, Yang K, Cuccarese M, Giedt RJ, Meimetis LG, et al. Quantitating drug-target engagement in single cells in vitro and in vivo. Nat Chem Biol. 2017;13(2):168–73.

    Article  CAS  Google Scholar 

  18. Thurber GM, Yang KS, Reiner T, Kohler RH, Sorger P, Mitchison T, et al. Single-cell and subcellular pharmacokinetic imaging allows insight into drug action in vivo. Nat Commun. 2013;4:1504.

    Article  Google Scholar 

  19. Carney B, Kossatz S, Lok BH, Schneeberger V, Gangangari KK, Pillarsetty NVK, et al. Target engagement imaging of PARP inhibitors in small-cell lung cancer. Nat Commun. 2018;9(1):176.

    Article  Google Scholar 

  20. Kossatz S, Brand C, Gutiontov S, Liu JT, Lee NY, Gonen M, et al. Detection and delineation of oral cancer with a PARP1 targeted optical imaging agent. Sci Rep. 2016;6:21371.

    Article  CAS  Google Scholar 

  21. Ossovskaya V, Koo IC, Kaldjian EP, Alvares C, Sherman BM. Upregulation of poly (ADP-ribose) polymerase-1 (PARP1) in triple-negative breast cancer and other primary human tumor types. Genes Cancer. 2010;1(8):812–21.

    Article  CAS  Google Scholar 

  22. Nosho K, Yamamoto H, Mikami M, Taniguchi H, Takahashi T, Adachi Y, et al. Overexpression of poly(ADP-ribose) polymerase-1 (PARP-1) in the early stage of colorectal carcinogenesis. Eur J Cancer. 2006;42(14):2374–81.

    Article  CAS  Google Scholar 

  23. Staibano S, Pepe S, Lo Muzio L, Somma P, Mascolo M, Argenziano G, et al. Poly(adenosine diphosphate-ribose) polymerase 1 expression in malignant melanomas from photoexposed areas of the head and neck region. Hum Pathol. 2005;36(7):724–31.

    Article  CAS  Google Scholar 

  24. Michels J, Adam J, Goubar A, Obrist F, Damotte D, Robin A, et al. Negative prognostic value of high levels of intracellular poly(ADP-ribose) in non-small cell lung cancer. Ann Oncol. 2015;26(12):2470–7.

    Article  CAS  Google Scholar 

  25. Kossatz S, Pirovano G, Demetrio De Souza Franca P, Strome AL, Sunny SP, Zanoni DK, et al. Validation of the use of a fluorescent PARP1 inhibitor for the detection of oral, oropharyngeal and oesophageal epithelial cancers. Nat Biomed Eng. 2020;4(3):272–85.

    Article  CAS  Google Scholar 

  26. Thurber GM, Reiner T, Yang KS, Kohler RH, Weissleder R. Effect of small-molecule modification on single-cell pharmacokinetics of PARP inhibitors. Mol Cancer Ther. 2014;13(4):986–95.

    Article  CAS  Google Scholar 

  27. Hernot S, van Manen L, Debie P, Mieog JSD, Vahrmeijer AL. Latest developments in molecular tracers for fluorescence image-guided cancer surgery. Lancet Oncol. 2019;20(7):e354–e67.

    Article  CAS  Google Scholar 

  28. Pirovano G, Roberts S, Kossatz S, Reiner T. Optical imaging modalities: principles and applications in preclinical research and clinical settings. J Nucl Med. 2020;61(10):1419–27. https://doi.org/10.2967/jnumed.119.238279.

    Article  CAS  PubMed  Google Scholar 

  29. Demétrio de Souza França P, Guru N, Roberts S, Kossatz S, Mason C, Abrahão M, Ghossein RA, Patel SG, Reiner T. Fluorescence-guided resection of tumors in mouse models of oral cancer. Sci Rep. 2020;10(1):11175. https://doi.org/10.1038/s41598-020-67958-8.

  30. Tang J, Salloum D, Carney B, Brand C, Kossatz S, Sadique A, et al. Targeted PET imaging strategy to differentiate malignant from inflamed lymph nodes in diffuse large B-cell lymphoma. Proc Natl Acad Sci U S A. 2017;114(36):E7441–e9.

    Article  CAS  Google Scholar 

  31. Kossatz S, Weber WA, Reiner T. Optical imaging of PARP1 in response to radiation in oral squamous cell carcinoma. PLoS One. 2016;11(1):e0147752.

    Article  Google Scholar 

  32. Demétrio de Souza Franca P, Guru N, Kostolansky AR, Mauguen A, Pirovano G, Kossatz S, Roberts S, Abrahao M, Patel SG, Reiner T, Jewell E. Poly(ADP-ribose)polymerase1: a potential molecular marker to identify cancer during colposcopy procedures. J Nucl Med. 2020. https://doi.org/10.2967/jnumed.120.253575

  33. Koch M, Symvoulidis P, Ntziachristos V. Tackling standardization in fluorescence molecular imaging. Nat Photonics. 2018;12(9):505–15.

    Article  CAS  Google Scholar 

  34. Kossatz S, Weber W, Reiner T. Detection and delineation of oral cancer with a PARP1-targeted optical imaging agent. Mol Imaging. 2017. https://doi.org/10.1177/1536012117723786.

  35. Carney B, Kossatz S, Reiner T. Molecular imaging of PARP. J Nucl Med. 2017;58(7):1025–30.

    Article  CAS  Google Scholar 

  36. van Dam GM, Themelis G, Crane LM, Harlaar NJ, Pleijhuis RG, Kelder W, et al. Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-alpha targeting: first in-human results. Nat Med. 2011;17(10):1315–9.

    Article  Google Scholar 

  37. Schebesch KM, Rosengarth K, Brawanski A, Proescholdt M, Wendl C, Höhne J, et al. Clinical benefits of combining different visualization modalities in neurosurgery. Front Surg. 2019;6:56.

    Article  Google Scholar 

Download references

Acknowledgements

The authors gratefully acknowledge the support of the Memorial Sloan Kettering Cancer Radiochemistry & Molecular Imaging Probes Core, the Molecular Cytology Core, and the Memorial Sloan Kettering Center for Molecular Imaging & Nanotechnology. We especially thank Eric Chan for the help with the ImageJ plugins for PARP1 and PARPi-FL quantifications. We also thank Garon Scott for proofreading and Terry Helms for creating illustrations. We also thank the operating room nurses and the Head and Neck surgical faculty and clinical staff at MSK, especially Violeta Dokic, R.N. for the help with the patients.

Availability of data and materials

All data and material are made available.

Funding

This work was supported by National Institutes of Health grants P30 CA008748, R01 CA204441 (TR), and R43 CA228815 (CB and TR). Dr. Valero was sponsored by a grant from Fundación Alfonso Martín Escudero. The funding sources were not involved in study design, data collection and analysis, writing of the report, or the decision to submit this article for publication.

Author information

Authors and Affiliations

Authors

Contributions

P.D.S.F., S.K., C.B; S.P., and T.R. conceived the study and designed the experiments. P.D.S.F, S.K., C.B, N.G., D.K.Z., D.A., C.V., R.G.G., H.S., I.G., S.P., and T.R. carried out the experiments and collected the data. C.B., S.R. and T.R. produced GMP PARPi-FL. P.D.S.F., C.B., R.A.G., S.P., and T.R. analyzed the data. P.D.S.F., S.K., C.B., N.G., W.A.W.; S.P., and T.R. wrote IRB protocols. P.D.S.F., C.B., A.M., S.P., and T.R. conducted statistical analysis of the data. P.D.S.F., S.K., C.B., and T.R. primarily wrote the manuscript.

Corresponding authors

Correspondence to Snehal G. Patel or Thomas Reiner.

Ethics declarations

Ethics approval

This study is compliant with the Health Insurance Portability and Accountability Act, was approved by the MSK Institutional Review Board, and was conducted per the Declaration of Helsinki ethical principles.

Consent for publication

All of the authors have read and approved the manuscript and possible conflict of interests are disclosed.

Conflict of interest

C.B., S.K., S.P., and T.R. are shareholders of Summit Biomedical Imaging, LLC. S.K., S.P., and T.R. are co-inventors on PCT application WO2016164771. T.R. is co-inventor on PCT application WO2012074840. T.R. is a paid consultant for Theragnostics, Inc. All the other authors have no relevant conflict to declare. This arrangement has been reviewed and approved by Memorial Sloan Kettering Cancer Center in accordance with its conflict of interest policies.

Key point question

Is the topical application of PARPi-FL safe? Can a mouthwash of the imaging agent provide a level of optical contrast (tumor to margin ratio) apparent to the human observer?

Pertinent findings

Topical application of PARPi-FL is safe and generated contrast-ratios above 3. A benign lesion did not take up the agent, and an undiagnosed contralateral small tumor was identified. PARPi-FL generates mesoscopic contrast by emitting a signal from its nuclear target on a subcellular level.

Implications for patient care

PARPi-FL is a simple, in vivo, non-invasive, cost-effective, molecularly specific, fluorescent contrast-based method to enhance oral cancer diagnosis.

Additional information

Publisher’s note

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

This article is part of the Topical Collection on Oncology - Head and Neck

Supplementary information

ESM 1

(DOCX 6918 kb)

ESM 2

(PDF 114 kb)

ESM 3

(PDF 4.56 mb)

ESM 4

(PDF 329 kb)

ESM 5

(PDF 727 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Demétrio de Souza França, P., Kossatz, S., Brand, C. et al. A phase I study of a PARP1-targeted topical fluorophore for the detection of oral cancer. Eur J Nucl Med Mol Imaging 48, 3618–3630 (2021). https://doi.org/10.1007/s00259-021-05372-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00259-021-05372-6

Keywords

Navigation