Abstract

Canine diffuse large B-cell lymphoma (DLBCL) is one of the most common cancers in dogs which shares remarkable similarities with its human counterpart, making the dog an excellent model for the investigation of novel therapeutic agents. However, the integration of canine lymphoma in comparative studies has been limited due in part to the lack of suitable xenograft mouse models for preclinical studies. To overcome these limitations, we established and characterized a localized subcutaneous bioluminescent canine DLBCL xenograft mouse model. The canine CLBL-1 cell line stably expressing the luciferase and green fluorescent protein reporters was generated and used to establish the xenograft tumor model. A pilot study was first conducted with three different cell densities (0.1×106, 0.5×106 and 1×106 cells) in SCID mice. All mice presented homogeneous tumor induction within eight days after subcutaneous injection, with a 100% engraftment efficiency and no significant differences were observed among groups. The tumors were highly aggressive and localized at the site of inoculation and reproduced histological features and immunophenotype consistent with canine DLBCL. Importantly, xenograft tumors were detected and quantified by bioluminescent imaging. To assess response to therapy, a therapeutic study with a histone deacetylase inhibitor, panobinostat, was performed. The results demonstrated that panobinostat (20 mg/kg) efficiently inhibited tumor growth and that bioluminescent imaging allowed the monitorization and quantification of tumor response to therapy. In summary, this study provides a bioluminescence canine DLBCL model that offers high engraftment efficiency, preservation of tumor features, and noninvasive monitoring of tumor progression, validating the model as a promising preclinical tool for both veterinary and human medicine.

Highlights

  • Non-Hodgkin lymphoma (NHL) is a leading cause of cancer-related death in the United States and Europe, and its incidence continues to increase [1,2]

  • The CLBL-1 cell line was selected for our study because it is the only canine cell line that faithfully represents diffuse large B-cell lymphoma (DLBCL), reproducibly inducing tumors and preserving its phenotype in the xenotransplantation setting [11,23,24,27]

  • The evaluation of growth patterns through a cell doubling time assay confirmed that the stable CLBL-1GFP+luciferase+ cell line exhibited a similar doubling time compared to the CLBL-1 parental cell line (26.45 hour doubling time for CLBL-1GFP+luciferase+, versus 26.52 hours for the parental CLBL-1 cell line) (S1 Fig)

Read more

Summary

Introduction

Non-Hodgkin lymphoma (NHL) is a leading cause of cancer-related death in the United States and Europe, and its incidence continues to increase [1,2]. Dogs diagnosed with lymphoma are frequently treated with anthracycline based chemotherapy regimens, to human DLBCL patients, providing realistic opportunities to explore therapeutic protocols that may translate to human clinical trials [8]. These initiatives are encouraged by the increasing healthcare standards demanded by pet owners, creating the need for novel cancer therapies designed for veterinary applications [11,12,13]. Even though comparative oncology studies provide unique information not acquired with conventional preclinical models, the use of the tumor-bearing dog model for innovative drug development requires previous controlled toxicokinetic studies in laboratory animals [14]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call