Development of a human immuno-oncology therapeutic agent targeting HER2: targeted delivery of granzyme B

J Exp Clin Cancer Res. 2019 Jul 30;38(1):332. doi: 10.1186/s13046-019-1333-6.

Abstract

Background: Immunotherapeutic approaches designed to augment T and B cell mediated killing of tumor cells has met with clinical success in recent years suggesting tremendous potential for treatment in a broad spectrum of tumor types. After complex recognition of target cells by T and B cells, delivery of the serine protease granzyme B (GrB) to tumor cells comprises the cytotoxic insult resulting in a well-characterized, multimodal apoptotic cascade.

Methods: We designed a recombinant fusion construct, GrB-Fc-4D5, composed of a humanized anti-HER2 scFv fused to active GrB for recognition of tumor cells and internal delivery of GrB, simulating T and B cell therapy. We assessed the construct's antigen-binding specificity and GrB enzymatic activity, as well as in vitro cytotoxicity and internalization into target and control cells. We also assessed pharmacokinetic and toxicology parameters in vivo.

Results: GrB-Fc-4D5 was highly cytotoxic to Her2 positive cells such as SKBR3, MCF7 and MDA-MB-231 with IC50 values of 56, 99 and 27 nM, respectively, and against a panel of HER2+ cell lines regardless of endogenous expression levels of the PI-9 inhibitor. Contemporaneous studies with Kadcyla demonstrated similar levels of in vitro activity against virtually all cells tested. GrB-Fc-4D5 internalized rapidly into target SKOV3 cells within 1 h of exposure rapidly delivering GrB to the cytoplasmic compartment. In keeping with its relatively high molecular weight (160 kDa), the construct demonstrated a terminal-phase serum half-life in mice of 39.2 h. Toxicity studies conducted on BALB/c mice demonstrated no statistically significant changes in SGPT, SGOT or serum LDH. Histopathologic analysis of tissues from treated mice demonstrated no drug-related changes in any tissues examined.

Conclusion: GrB-Fc-4D5 shows excellent, specific cytotoxicity and demonstrates no significant toxicity in normal, antigen-negative murine models. This construct constitutes a novel approach against HER2-expressing tumors and is an excellent candidate for further development.

Keywords: Granzyme B; HER2; Immunotherapy; Kadcyla; Pharmacokinetics.

MeSH terms

  • Animals
  • Antigens, Neoplasm / immunology
  • Antineoplastic Agents, Immunological / isolation & purification
  • Antineoplastic Agents, Immunological / pharmacology*
  • Cell Line, Tumor
  • Cell Survival / drug effects
  • Disease Models, Animal
  • Drug Delivery Systems
  • Drug Development*
  • Gene Expression
  • Genetic Vectors / genetics
  • Granzymes / administration & dosage
  • Granzymes / genetics
  • Humans
  • Mice
  • Molecular Targeted Therapy* / methods
  • Protein Binding / immunology
  • Receptor, ErbB-2 / antagonists & inhibitors*
  • Recombinant Fusion Proteins / administration & dosage
  • Recombinant Fusion Proteins / genetics
  • Recombinant Fusion Proteins / isolation & purification
  • Recombinant Fusion Proteins / pharmacology*
  • Single-Chain Antibodies / genetics
  • Xenograft Model Antitumor Assays

Substances

  • Antigens, Neoplasm
  • Antineoplastic Agents, Immunological
  • Recombinant Fusion Proteins
  • Single-Chain Antibodies
  • ERBB2 protein, human
  • Receptor, ErbB-2
  • GZMB protein, human
  • Granzymes