2,263
Views
1
CrossRef citations to date
0
Altmetric
Report

Development and pharmacokinetic assessment of a fully canine anti-PD-1 monoclonal antibody for comparative translational research in dogs with spontaneous tumors

, , , , , , , , & ORCID Icon show all
Article: 2287250 | Received 11 Jul 2023, Accepted 20 Nov 2023, Published online: 04 Dec 2023
 

ABSTRACT

PD-1 checkpoint inhibitors have revolutionized the treatment of patients with different cancer histologies including melanoma, renal cell carcinoma, and non-small cell lung carcinoma. However, only a subset of patients show a dramatic clinical response to treatment. Despite intense biomarker discovery efforts, no single robust, prognostic correlation has emerged as a valid outcome predictor. Immune competent, pet dogs develop spontaneous tumors that share similar features to human cancers including chromosome aberrations, molecular subtypes, immune signatures, tumor heterogeneity, metastatic behavior, and chemotherapeutic response. As such, they represent a valuable parallel patient population in which to investigate predictive biomarkers of checkpoint inhibition. However, the lack of a validated, non-immunogenic, canine anti-PD-1 antibody for pre-clinical use hinders this comparative approach and prevents potential clinical benefits of PD-1 blockade being realized in the veterinary clinic. To address this, fully canine single-chain variable fragments (scFvs) that bind canine (c)PD-1 were isolated from a comprehensive canine scFv phage display library. Lead candidates were identified that bound with high affinity to cPD-1 and inhibited its interaction with canine PD-L1 (cPD-L1). The lead scFv candidate re-formatted into a fully canine IgGD reversed the inhibitory effects of cPD-1:cPD-L1 interaction on canine chimeric antigen receptor (CAR) T cell function. In vivo administration showed no toxicity and revealed favorable pharmacokinetics for a reasonable dosing schedule. These results pave the way for clinical trials with anti-cPD-1 in canine cancer patients to investigate predictive biomarkers and combination regimens to inform human clinical trials and bring a promising checkpoint inhibitor into the veterinary armamentarium.

Abbreviations

ADA=

anti-drug antibodies

AE=

adverse events

AUCinf=

area under the serum concentration vs. time curve extrapolated to time infinity

AUClast=

area under the serum concentration vs. time curve from time 0 to 21 days

CAR=

chimeric antigen receptor

CL=

clearance

Cmax=

maximum observed serum concentration

CMS=

carboxymethyl surface

cPBMCs=

canine peripheral blood mononuclear cells

CTLA4=

cytotoxic T lymphocyte antigen 4

CTV=

cell trace violet

DCs=

dendritic cells

ELISA=

enzyme linked immunosorbent assay

FDA=

Federal Drug Administration

GLP=

good laboratory practice

HA=

hemagglutinin

HIS=

6x histidine tag

HNSTD=

highest non severely toxic dose

IACUC=

institutional animal care and use committee

ICI=

immune checkpoint inhibitor

NCA=

non-compartmental analysis

NOAEL=

No Observed Adverse Event Level

LAG3=

lymphocyte-activation gene 3

MERS=

Middle Eastern Respiratory Syndrome

MFI=

mean fluorescence intensity

MSI-H/MMRD=

microsatellite instability-high/mis-match repair deficiency

MTD=

maximum tolerated dose

PALS=

periarteriolar lymphoid sheaths

PD-1=

programmed cell death protein 1

PD-L1=

programmed cell death ligand 1

PK=

pharmacokinetics

RO=

receptor occupancy

scFv=

single chain variable fragment

SDS-PAGE=

Sodium dodecyl-sulfate polyacrylamide gel electrophoresis

SEC-HPLC=

Size exclusion-high-performance liquid chromatography

t1/2=

terminal log-linear half-life

TIGIT=

T cell immunoreceptor with immunoglobulin and ITIM domain

TILs=

tumor infiltrating lymphocytes

TIM-3=

T cell immunoglobulin and mucin-domain containing-3

TMB=

tumor mutational burden

TMDD=

Target Mediated Drug Disposition

VH=

variable heavy

VL=

variable light

Vss=

volume of distribution 

Acknowledgments

We would like to thank Joel Cassel and the Wistar Molecular Screening and Protein Expression facility for their assistance and expertise with SPR performance and analysis. We are also grateful for the support of the Penn Vet Comparative Pathology Core, which is part of the Abramson Cancer Center Support Grant (P30 CA016520); the Aperio Versa 200 scanner used for imaging was acquired through an NIH Shared Instrumentation Grant (S10 OD023465-01A1); the Leica BOND RXm instrument used for IHC and ISH was acquired through the Penn Vet IIZD Core pilot grant opportunity 2022. Special thanks to Amiko Saito Yoshimoto for assistance with high-resolution figures.

Disclosure statement

Authors NJM, DLS, HW, and MB have equity in Vetigenics LLC.

Supplementary material

Supplemental data for this article can be accessed online at https://doi.org/10.1080/19420862.2023.2287250.

Additional information

Funding

This work was supported by the V Foundation (NJM & DS), Vetigenics LLC, a generous gift from Mr. and Mrs. D. Sabey, and the NCI/SBIR under Contract 75N91018C000042 (Vetigenics LLC (NC)). SY is supported by the Japan Society for the Promotion of Science (JSPS) KAKENHI program grant numbers JP21J01155 and JP22K15018. PG is supported by NCI/NHLBI HL153696.