Kharkwal et al. showed that covalent conjugation of the CD1d agonist, α-galactosylceramide (α-GalCer), to a soluble CD1d/Β2m fusion protein linked to mouse or human HER2-specific ScFvs enabled durable iNKT cell activation. In vitro and in wild-type, CD1d-/-, or human CD1d knock-in mice, α-GalCer/HER2 BiTEs directly stimulated iNKT cells to induce DC maturation, NK and T cell activation, and epitope spreading for tumor-specific CD8+ T cell responses. BiTEs regressed grafted human Her2+ metastatic melanoma and colorectal tumor lines, synergized with anti-CTLA-4, and induced tumor-specific recall responses. Repeated BiTE injection did not induce anergy.

Contributed by Paula Hochman

ABSTRACT: CD1d-restricted invariant natural killer T cells (iNKT cells) mediate strong anti-tumor immunity when stimulated by glycolipid agonists. However, attempts to develop effective iNKT cell agonists for clinical applications have been thwarted by potential problems with dose-limiting toxicity and by activation-induced iNKT cell anergy, which limits the efficacy of repeated administration. To overcome these issues, we developed a unique bispecific T cell engager (BiTE) based on covalent conjugates of soluble CD1d with photoreactive analogs of the glycolipid α-galactosylceramide. Here we characterize the in vivo activities of iNKT cell-specific BiTEs and assess their efficacy for cancer immunotherapy in mouse models using transplantable colorectal cancer or melanoma tumor lines engineered to express human Her2 as a tumor-associated antigen. Systemic administration of conjugated BiTEs stimulated multiple iNKT cell effector functions including cytokine release, secondary activation of NK cells, and induction of dendritic cell maturation and also initiated epitope spreading for tumor-specific CD8+ cytolytic T cell responses. The anti-tumor effects of iNKT cell activation with conjugated BiTEs were further enhanced by simultaneous checkpoint blockade with antibodies to CTLA-4, providing a potential approach for combination immunotherapy. Multiple injections of covalently stabilized iNKT cell-specific BiTEs activated iNKT cells without causing iNKT anergy and exhaustion, thus enabling repeated administration for effective and nontoxic cancer immunotherapy regimens.

Author Info: (1) Microbiology & Immunology, Albert Einstein College of Medicine. (2) Microbiology & Immunology, Albert Einstein College of Medicine. (3) School of Biosciences, University of Bir

Author Info: (1) Microbiology & Immunology, Albert Einstein College of Medicine. (2) Microbiology & Immunology, Albert Einstein College of Medicine. (3) School of Biosciences, University of Birmingham. (4) Albert Einstein College of Medicine. (5) Microbiology & Immunology, Albert Einstein College of Medicine. (6) Millenium Institute on Immunology & Immunotherapy, Programa de Inmunologia, Instituto de Ciencias Biomedicas, Facultad de Medicina, Universidad de Chile. (7) Microbiology & Immunology, Albert Einstein College of Medicine. (8) Microbiology & Immunology, Albert Einstein College of Medicine. (9) Biochemistry, Albert Einstein College of Medicine. (10) Centre of Chemistry, University of Minho. (11) Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences/Peking Union Medical College. (12) Ludwig Institute for Cancer Research, University of Lausanne. (13) School of Biosciences, University of Birmingham. (14) School of Chemistry, University of Birmingham. (15) Biochemistry, Albert Einstein College of Medicine. (16) Research, Vaccinex Inc. (17) Research, Vaccinex Inc. (18) Research, Vaccinex, Inc. (19) University of Birmingham. (20) Microbiology & Immunology, Albert Einstein College of Medicine steven.porcelli@einsteinmed.org.