Abstract

BackgroundImmunotherapeutic 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.MethodsWe 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.ResultsGrB-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.ConclusionGrB-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.

Highlights

  • 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

  • The current study extends our initial observations of impressive biological activity of granzyme B (GrB)-containing anti-HER2/ neu fusion constructs and provides a comparison to the FDA-approved Antibody drug conjugate (ADC) Kadcyla

  • Construction, expression, and purification of GrB-Fc-Humanized anti-HER2/neu scFv (4D5) fusion protein Following expression and protein purification through Nickel-IMAC, the fusion protein was incubated with recombinant enterokinase to remove the N-terminal purification tag

Read more

Summary

Introduction

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. GrB has been well validated as a highly cytotoxic payload with a unique mechanism of action when compared to other payloads employed in targeted therapeutic constructs including antibody drug conjugates (ADCs) and fusion toxins [27,28,29,30,31,32]. The addition of a highly cytotoxic payload emtansine to Herceptin has resulted in an agent Kadcyla (T-DM1) with excellent clinical therapeutic properties [42,43,44,45], as well as a number of follow-on ADC products for a wide-range of therapeutic targets, including new designs and novel payloads with unique mechanisms of action [46,47,48,49,50]. The emergence of resistance mechanisms which limit treatment success with these agents is driving innovation to overcome these issues

Methods
Results
Discussion
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