Observing that monocytes from the ascites fluid of murine i.p. ovarian tumors carried tumor antigens, Adams and Grimm et al. showed that although these monocytes were unable to effectively stimulate T cell responses, a 48 hour ex vivo stimulation with TLR4 and TLR9 ligands and blockade of immunosuppressive IL-10 enhanced their APC phenotype and stimulated autologous tumor-derived T cells. Vaccination with activated monocytes prevented and controlled ID8 ovarian tumors, extended survival, and induced both CD4+ and CD8+ antitumor memory. Monocytes from human ascites samples effectively presented tumor-derived TAAs.

Contributed by Ed Fritsch

BACKGROUND: Novel therapeutic strategies in ovarian cancer (OC) are needed as the survival rate remains dismally low. Although dendritic cell-based cancer vaccines are effective in eliciting therapeutic responses, their complex and costly manufacturing process hampers their full clinical utility outside specialized clinics. Here, we describe a novel approach of generating a rapid and effective cancer vaccine using ascites-derived monocytes for treating OC. METHODS: Using the ID8 mouse ovarian tumor model and OC patient samples, we isolated ascites monocytes and evaluated them with flow cytometry, Luminex cytokine and chemokine array analysis, ex vivo cocultures with T cells, in vivo tumor challenge and T cell transfer experiments, RNA-sequencing and mass spectrometry. RESULTS: We demonstrated the feasibility of isolating ascites monocytes and restoring their ability to function as bona fide antigen-presenting cells (APCs) with Toll-like receptor (TLR) 4 lipopolysaccharide and TLR9 CpG-oligonucleotides, and a blocking antibody to interleukin-10 receptor (IL-10R Ab) in the ID8 model. The ascites monocytes were laden with tumor antigens at a steady state in vivo. After a short 48_hours activation, they upregulated maturation markers (CD80, CD86 and MHC class I) and demonstrated strong ex vivo T cell stimulatory potential and effectively suppressed tumor and malignant ascites in vivo. They also induced protective long-term T cell memory responses. To evaluate the translational potential of this approach, we isolated ascites monocytes from stage III/IV chemotherapy-nave OC patients. Similarly, the human ascites monocytes presented tumor-associated antigens (TAAs), including MUC1, ERBB2, mesothelin, MAGE, PRAME, GPC3, PMEL and TP53 at a steady state. After a 48-hour treatment with TLR4 and IL-10R Ab, they efficiently stimulated oligoclonal tumor-associated lymphocytes (TALs) with strong reactivity against TAAs. Importantly, the activated ascites monocytes retained their ability to activate TALs in the presence of ascitic fluid. CONCLUSIONS: Ascites monocytes are naturally loaded with tumor antigen and can perform as potent APCs following short ex vivo activation. This novel ascites APC vaccine can be rapidly prepared in 48_hours with a straightforward and affordable manufacturing process, and would be an attractive therapeutic vaccine for OC.

Author Info: (1) Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA. Division of Gynecologic Oncology, University of New Mexico Comprehensive Cancer Cen

Author Info: (1) Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA. Division of Gynecologic Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA. (2) Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (3) Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (4) Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA. (5) Division of Gynecologic Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA. (6) Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA. (7) Division of Gynecologic Oncology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA. (8) Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (9) Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (10) Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (11) Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. (12) Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (13) Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (14) Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (15) Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA. (16) Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. Ludwig Institute for Cancer Research, Lausanne, Switzerland. (17) Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA. (18) Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland Lana.Kandalaft@chuv.ch. Ludwig Institute for Cancer Research, Lausanne, Switzerland.