Invariant natural killer T cells (iNKT) are able to recognize and kill cells presenting the lipid agonist αGC on CD1d. To exploit iNKT cells in a CD1d-independent manner, Das et al. developed a soluble bispecific fusion protein linking the CD1d molecule to a CD19 scFv. When loaded with αGC, the fusion protein engaged iNKT cells in vitro, inducing activation, cytokine production, proliferation, and specific killing of CD19+CD1d- target cells, including EBV-immortalized B cells. Activated iNKT cells promoted DC maturation, driving the activation of a broader immune response. In vivo, the fusion protein controlled CD19+CD1d- tumor growth.

Invariant natural killer T (iNKT) cells comprise a unique lineage of CD1d-restricted lipid-reactive T lymphocytes that potently kill tumor cells and exhibit robust immunostimulatory functions. Optimal tumor-directed iNKT cell responses often require expression of the antigen-presenting molecule CD1d on tumors; however, many tumor cells downregulate CD1d and thus evade iNKT cell recognition. We generated a soluble bispecific fusion protein designed to direct iNKT cells to the site of B-cell cancers in a tumor antigen-specific but CD1d-independent manner. This fusion protein is composed of a human CD1d molecule joined to a single chain antibody FV fragment specific for CD19, an antigen widely expressed on B-cell cancers. The CD1d-CD19 fusion protein binds specifically to CD19-expressing, but not CD19-negative cells. Once loaded with the iNKT cell lipid agonist alpha-galactosyl ceramide (alphaGC), the CD1d-CD19 fusion induces robust in vitro activation of and cytokine production by human iNKT cells. iNKT cells stimulated by the alphaGC-loaded CD1d-CD19 fusion also strongly transactivate T-, B-, and NK-cell responses and promote dendritic cell maturation. Importantly, the alphaGC-loaded fusion induces robust lysis of CD19(+)CD1d(-) Epstein-Barr virus immortalized human B-lymphoblastoid cell lines that are otherwise resistant to iNKT cell killing. Consistent with these findings; administration of the alphaGC-loaded fusion protein controlled the growth of CD19(+)CD1d(-) tumors in vivo, suggesting that it can "link" iNKT cells and CD19(+)CD1d(-) targets in a therapeutically beneficial manner. Taken together, these preclinical studies demonstrate that this B cell-directed fusion protein can be used to effectively induce iNKT cell antitumor responses in vitro and in vivo.

Author Info: (1) Department of Physiology, Michigan State University, East Lansing, MI. (2) Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA. (3) Division of Oncology

Author Info: (1) Department of Physiology, Michigan State University, East Lansing, MI. (2) Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA. (3) Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA. (4) Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA. (5) Department of Physiology, Michigan State University, East Lansing, MI. (6) Vaccinex Inc., Rochester, NY; and. (7) Vaccinex Inc., Rochester, NY; and. (8) Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN.