Elsevier

Biomaterials

Volume 163, May 2018, Pages 67-75
Biomaterials

STINGel: Controlled release of a cyclic dinucleotide for enhanced cancer immunotherapy

https://doi.org/10.1016/j.biomaterials.2018.01.035Get rights and content

Abstract

Recent advancements in the field of immunotherapy have yielded encouraging results for the treatment of advanced cancers. Cyclic dinucleotides (CDNs) are a powerful new class of immunotherapy drugs known as STING (Stimulator of Interferon Genes) agonists, currently in clinical trials. However, previous studies of CDNs in murine cancer models have required multiple injections, and improve survival only in relatively nonaggressive tumor models. Therefore, we sought to improve the efficacy of CDN immunotherapy by developing a novel biomaterial we call “STINGel.” STINGel is an injectable peptide hydrogel that localizes and provides controlled release of CDN delivery, showing an 8-fold slower release rate compared to a standard collagen hydrogel. The carrier hydrogel is a positively charged, MultiDomain Peptide (MDP) which self-assembles to form a nanofibrous matrix and is easily delivered by syringe. The highly localized delivery of CDN from this nanostructured biomaterial affects the local histological response in a subcutaneous model, and dramatically improves overall survival in a challenging murine model of head and neck cancer compared to CDN alone or CDN delivered from a collagen hydrogel. This study demonstrates the feasibility of biomaterial-based immunotherapy platforms like STINGel as strategies for increasing the efficacy of CDN immunotherapies.

Introduction

Essential to modern medicine is the effective delivery of drug therapies. Injectable biomaterials have been established as powerful methods of therapy administration due to their ability to control the release profile of loaded agents and influence the local biological reponse [[1], [2], [3]]. Such biomaterial-based delivery systems have been used for a host of useful applications, including tissue engineering, growth factor release, DNA delivery, and vaccine incorporation [[4], [5], [6], [7]]. Recent interest has focused on biomaterials as platforms for cancer immunotherapy, which can provide extended and localized drug release, and also allow for the intelligent modulation of immune cells in situ.

Cancer immunotherapy has arisen as an exciting treatment modality for several advanced-stage cancers, with the potential to generate specific and durable anti-tumor responses through the use of the host's natural immune reponse [[8], [9], [10], [11]]. Significant progress has been made in this field, generating encouraging results in the treatment of cancers such as metastatic melanoma, for which the 5-year overall survival rate has increased from less than 10%, to almost 40% [[12], [13], [14]]. However, in the case of other types of cancer such as head and neck squamous cell carcinoma (HNSCC) survival rates have failed to improve at a comparable rate, remaining relatively stagnant for the past few decades and emphasizing the need for new treatment options [15].

HNSCC is the sixth most common cancer worldwide, with more than 60,000 men and women in the U.S. expected to develop HNSCC this year [16,17]. Etiologic risk factors include exposure to carcinogens such as tobacco and alcohol, and viral infections such as high-risk types of human papilloma viruses (HPVs) [[18], [19], [20]]. Given the well-known co-morbidities and recurrence rates associated with conventional treatments such as surgery, radiation therapy, and chemotherapy, there is a real need for innovative approaches to treat HNSCC. While some HNSCC patients undergoing immune checkpoint inhibitor antibody therapy experience durable complete remissions, the majority (80–85%) of patients derive no benefit [21]. Improving the rate of success in HNSCC immunotherapy is therefore a highly desirable goal.

Recently, a novel class of immunotherapeutics based on synthetic cyclic dinucleotides (CDNs) have been found to induce strong anti-tumor responses in preclinical models through the Stimulator of Interferon Genes (STING) pathway [[22], [23], [24]]. The STING pathway has emerged as a key mechanism linking the detection of cytosolic tumor DNA to downstream activation of innate immune cells [25,26]. The rationally-designed synthetic Cyclic Dinucleotide dithio-(RP,RP)-[cyclic[A(2′,5′)pA(3′,5′)p]] (abbreviated as ML RR-S2 CDA or just CDN, see Fig. 1A) is a promising candidate molecule in clinical trials (see NCT02675439) that has shown efficacy in murine cancer models, promoting the specific rejection of several types of tumors [[22], [27]]. However, to date CDN monotherapy has shown poor efficacy in preclinical models of HNSCC, requiring multiple injections and concomitant administration of immune checkpoint antibodies [28]. Current clinical trials are evaluating intratumoral injections of CDN as monotherapy, a strategy that may prove to be insufficient [27]. Thus, novel approaches to improve the efficacy of CDN in challenging, treatment-refractory tumor models are warranted.

In response to this challenge, we have developed a novel peptide hydrogel-based platform for intratumoral CDN delivery, which we call “STINGel.” This localizable drug delivery vehicle, utilizing the power of immunotherapy, is based on prior work in our laboratory studying the utility of multidomain peptides (MDPs) as unique supramolecular biomaterials. These self-assembling peptides mimic the extracellular matrix of cells through the formation of a nanofibrous network, and can act as biofunctional delivery platforms that allow for an immense diversity of functionality to be introduced [29,30]. Rationally engineered MDPs have been shown to form anti-parallel β-sheets of peptide nanofibers in solution, which when electrostatically crosslinked with multivalent ions form extended nanofiber networks to create robust hydrogels (see Fig. 1B–D) [30].

MDPs possess a number of attractive characteristics as biomaterials. 1) The peptide hydrogels are thixotropic, allowing them to be easily delivered by syringe and yet remain localized for applications such as intratumoral injections. 2) The design of the peptide sequence can allow for the incorporation of charged small molecule drugs by nanofiber crosslinking to achieve extended release, or can allow for the encapsulation and gradual release of small hydrophobic drugs such as daunorubicin, etodolac, levofloxacin, and others [29,31,32]. 3) The hydrogels undergo complete cellular infiltration within three days post injection in vivo and are not fibrously encapsulated, maximizing matrix-tissue interaction.

In this study, we sought to use the MDP hydrogel with sequence K2(SL)6K2 to deliver a promising STING agonist CDN, by taking advantage of favorable electrostatic interactions between the CDN's negative thiophosphate linkages and the positive lysine residues at the peptide termini (Fig. 1). We hypothesized that a combination of controlled release and favorable local environment would result in improved tumor treatment efficacy in vivo.

Section snippets

Peptide synthesis

Peptide synthesis reagents were purchased from EMD Chemicals (Philadelphia, PA). An Apex Focus XC (Aapptec) synthesizer was used to synthesize the multidomain peptide K2(SL)6K2 (MW = 1773.171 g/mol) according to a standard synthetic method previously published to yield pure peptide with acetylated N-terminus and C-terminal amide [30,32,33]. All peptides were analyzed by Autoflex MALDI-TOF MS (Bruker Instruments, Billerica, MA) for purity and confirmation of successful synthesis (Fig. S1).

Hydrogel preparation and loading

All

Hydrogel properties and drug release kinetics

The MDP hydrogel K2(SL)6K2, extensively characterized in past studies in terms of peptide secondary structure, nanostructure, rheological properties, and biocompatibility, was examined in the context of this experiment for its ability to encapsulate and deliver the small molecule CDN [[29], [30], [31]]. Ionic crosslinking of MDP fibers to achieve drug loading was previously accomplished with the negatively charged anti-parasitic compound suramin, and thus our work with CDN was a natural

Discussion

The ability of biomaterials to allow for spatiotemporal control over payload delivery means diverse factors can be released in a controlled manner within a specific volume, reducing off-target toxicity while also enabling localized improvements in efficacy. Materials able to exploit such capabilities to release factors that can intelligently direct and modulate immune cells in situ are highly attractive, and thus we sought to develop STINGel as such a platform to improve current immunotherapies.

Conclusions

We have developed a cyclic dinucleotide-loaded multidomain peptide hydrogel we call STINGel, which dramatically improves overall survival in a challenging murine oral cancer model compared to CDN monotherapy injection. In addition to a six-fold improvement in survival, 100% of surviving mice demonstrate immunological memory and reject a secondary challenge of cancer cells. STINGel shows extended release kinetics compared to a CDN loaded collagen gel. This translates into superior survival

Author contributions

DGL and JDH designed the kinetics, in vitro viability, and histological studies and DGL conducted the studies. TLLS provided additional histological data and interpretation. YLL provided pathological assessment of the histology data. ND, SLP, JDH, and SY designed the in vivo studies, and ND and SLP conducted the studies. DGL and ND analyzed the data, and DGL, JDH, ND, and SY interpreted the data. DGL, JDH, ND and SY wrote the manuscript.

Acknowledgments

This work was supported by the National Institutes of Health (grants DE021798, DE023577 and DE024173), the Oral and Maxillofacial Surgery Foundation (Research Support Grant), and the Welch Foundation (C1557). DGL was supported by the NSF Graduate Research Fellowship Program under Grant No. 1450681. TLLS was supported by the Mexican National Council for Science and Technology (CONACyT) PhD Scholarship Program. The authors would also like to thank Nicole Carrejo, and I-Che Li for scientific

References (45)

  • R. Langer et al.

    Tissue engineering

    Science

    (1993)
  • L.D. Shea et al.

    DNA delivery from polymer matrices for tissue engineering

    Nat. Biotechnol.

    (1999)
  • O.A. Ali et al.

    Infection-mimicking materials to program dendritic cells in situ

    Nat. Mater.

    (2009)
  • A. Modena et al.

    Immune checkpoint inhibitors and prostate cancer: a new Frontier?

    Oncol Rev.

    (2016)
  • K. Margolin

    The promise of molecularly targeted and immunotherapy for advanced melanoma

    Curr. Treat. Options Oncol.

    (2016)
  • P. Economopoulou et al.

    Checkpoint inhibitors in head and neck cancer: rationale, clinical activity, and potential biomarkers

    Curr. Treat. Options Oncol.

    (2016)
  • M.B. Atkins et al.

    Immunotherapy combined or sequenced with targeted therapy in the treatment of solid tumors: current perspectives

    JNCI (J. Natl. Cancer Inst.)

    (2016)
  • F. Hodi et al.

    Durable, long-term survival in previously treated patients with advanced melanoma (MEL) who received nivolumab (NIVO) monotherapy in a phase I trial: abstract CT001

  • A. Errico

    Melanoma: CheckMate 067–frontline nivolumab improves PFS alone or in combination with ipilimumab

    Nat. Rev. Clin. Oncol.

    (2015)
  • A. Hauschild et al.

    Combined immunotherapy–a new standard in metastatic melanoma?

    Nat. Rev. Clin. Oncol.

    (2015)
  • SEER Surveillance Epidemiology and End Results Fast Facts [Internet]....
  • SEER Cancer of the Oral Cavity and Pharynx - SEER Fast Fact Sheets. http://seer.cancer.gov/statfacts/html/oralcav.html...
  • Cited by (154)

    View all citing articles on Scopus
    1

    DGL and ND contributed equally.

    View full text