Elsevier

Journal of Controlled Release

Volume 309, 10 September 2019, Pages 173-180
Journal of Controlled Release

Inhibiting PI3 kinase-γ in both myeloid and plasma cells remodels the suppressive tumor microenvironment in desmoplastic tumors

https://doi.org/10.1016/j.jconrel.2019.07.039Get rights and content

Abstract

Phosphoinositide-3-kinases (PI3Ks) are part of signal transducing enzymes that mediate key cellular functions in cancer and immunity. PI3K-γ is crucial for cellular activation and migration in response to certain chemokines. PI3K-γ is highly expressed in myeloid cells and promotes their migration and the production of inflammatory mediators. We found that PI3K-γ was also highly expressed in tumor-associated B cells. IPI-549, the only PI3K-γ inhibitor in clinical development, offers a unique approach to enhance the anti-tumor immune response. We encapsulated IPI-549 in targeted polymeric nanoparticles (NP) and tested its activity in both murine pancreatic cancer and melanoma models. IPI-549 NP significantly decreased tumor growth and prolonged host survival in both models. Importantly, IPI-549 NP treatment reduced the suppressive tumor microenvironment by decreasing both suppressive myeloid and plasma cells in the tumor. We concluded that IPI-549 NP delivery could be a promising method for treating pancreatic cancer and other immune-suppressive tumors.

Introduction

Pancreatic cancer is a malignant disease with a high mortality rate. The total number of deaths from pancreatic cancer is steadily increasing, with pancreatic cancer expected to be the second leading cause of cancer death in the USA by 2030 [45]. Historically, chemotherapy or radiotherapy did not reach satisfactory survival benefit in advanced pancreatic cancer. Recent studies have revealed that immunosuppression and inflammation are related to oncogenesis, as well as tumor development, invasion, and metastasis in pancreatic adenocarcinoma (PAC). Therefore, immunosuppression associated signaling, especially when it involves immune checkpoint and inflammation, has served as a novel treatment target for PAC. There is a highly immunosuppressive microenvironment regulated by immune cells, stromal cells, and mediators in PAC. This condition may result in its resistance to immune checkpoint therapy [24].

Tumor-associated macrophages and myeloid-derived suppressor cells (MDSC) are immunosuppressive, and a high ratio of suppressive cells to CD4+ and CD8+ T cells is related to poor survival of PAC patients [24]. MDSC levels are increased both in the circulation and in the microenvironment of PAC. Inhibition of MDSC in PAC is a potential method of cancer therapy [51]. Phosphoinositide-3-kinases (PI3Ks) pertain to signal transducing enzymes that play an important role in cancer and immunity. PI3K-γ signaling is especially pivotal for the function of myeloid cells, where it is downstream of G-protein coupled receptors (GPCRs) (e.g., chemokine receptors) and RAS [22,49,50]. For instance, murine syngeneic tumors grow slower when transplanted into immune-competent mice where PI3K-γ is genetically inactivated [25,50]. This growth reduction occurs because of the abrogation of tumor-associated myeloid cells that are known to promote an immune-suppressive tumor microenvironment (TME) that permits tumor growth [18,25,46,50]. In addition, MDSCs are associated with tumor regrowth after radiation or chemotherapy, and are known to lead to metastatic spread [8]. These preclinical studies highlight an important role for PI3K-γ in myeloid cell biology and suggest that PI3K-γ inhibition in MDSC may be effective at inhibiting tumor growth in a variety of settings.

IPI-549 can reduce the T-cell-suppressive activity of both murine and human myeloid-derived suppressor cells in vitro [6]. These findings indicate that IPI-549 increases antitumor immunity by remodeling the tumor-immune microenvironment via blockade of tumor-associated myeloid cells. In addition, the up-regulations of costimulatory and coinhibitory genes with IPI-549 treatment provides a mechanistic rationale for the observed combination activity with immune checkpoint inhibition. IPI-549 is currently in phase I development, both as a single agent and in combination with an anti-PD-1 antibody, in solid tumors [40].

B cells have been typical of positive regulators of humoral immune response and are characterized by their ability to terminally differentiate into antibody-secreting plasma cells [9,33]. B-cell inhibition of an immune response was first reported in 1974; spleen B-cells were found to impair delayed type hypersensitivity response in guinea pigs [30,41]. B cells are now regarded as an important ingredient of the immune suppression system. A potential therapeutic strategy for PAC would include targeted B-cell suppression [18,35,44,47].

Nanoparticles (NPs) can prolong the half-life of payloads in vivo and passively accumulate in the tumor regions via the enhanced permeability and retention (EPR) effect [19,39,57]. Numerous anti-tumor or immune-stimulating agents have been delivered by NPs [6,58]. For example, a Toll-like-receptor-7 agonist was encapsulated by poly(lactic-co-glycolic) acid (PLGA) [28]. In the current study, targeted IPI-549 NP was designed to improve the bioavailability and therapeutic activity. We hypothesized that tumor-targeted IPI-549 could reshape the tumor immune microenvironment and reverse its immune suppression more effectively than the oral dosage form.

Section snippets

Materials

IPI-549 was purchased from Chemietek (Indianapolis, IN, USA). Acid-terminated PLGA (lactide/glycolide (50:50)) was purchased from DURECT (Pelham, AL, USA). mPEG3500-NH2·HCl and tBOC-PEG3500-NH2·HCl were purchased from JenKem Technology USA, Inc. (Allen, TX). PLGA-PEG and PLGA-PEG-AEAA were synthesized according to our previous work, and the structures were confirmed by 1H NMR [20]. If not specifically mentioned, all other chemicals were purchased from Sigma-Aldrich (St. Louis, MO, USA).

Cell culture

The

PI3K-γ in B cells

During inflammation and cancer, PI3K-γ controls a critical switch between immune stimulation and suppression. Previous studies have shown that PI3K-γ is highly expressed in myeloid cells [29]. As shown in Fig. 1, we demonstrated that PI3K-γ was also highly expressed in B cells associated with both KPC pancreatic cancer and BPD6 melanoma models. Both tumor models contained extensive stroma structure (Fig. S1), which is typical of an immune-suppressive microenvironment [37]. BRAF mutation is very

Discussion

Breg cells produce large amounts of cytokines such as IL-10, TGF-β, and IL-35, which suppress the differentiation of pro-inflammatory lymphocytes, such as tumor necrosis factor α (TNF-α)-producing monocytes, IL-12-producing dendritic cells, Th17 cells, Th 1 cells, and cytotoxic CD8+ T cells [36,53,55]. Breg cells can also result in the differentiation of immune-suppressive T cells, Foxp3+ T cells, and T regulatory 1 (Tr1) cells [3,12]. According to previous studies [6,29], PI3K-γ is highly

Author contributions

XZ, LS and LH (Huang) designed the study. XZ, LS, QL and LH (Hou) performed experiments and analyzed the data. XZ, LS and LH (Huang) wrote the manuscript, with help of QL for revisions. QL designed the graphical abstract.

Acknowledgments

This work was supported by NIH grant CA198999 and Eshelman Institute for Innovation.

References (59)

  • L.B. Rivera et al.

    Intratumoral myeloid cells regulate responsiveness and resistance to antiangiogenic therapy

    Cell Rep.

    (2015)
  • M.C. Schmid

    Receptor tyrosine kinases and TLR/IL1Rs unexpectedly activate myeloid cell PI3Kγ, a single convergent point promoting tumor inflammation and progression

    Cancer Cell

    (2011)
  • C.M. Sun et al.

    Upon TLR9 signaling, CD5+ B cells control the IL-12-dependent Th1-priming capacity of neonatal DCs

    Immunity

    (2005)
  • J. Zhang et al.

    Non-viral nanocarriers for siRNA delivery in breast cancer

    J. Control. Release

    (2014)
  • N.A. Carter et al.

    Mice lacking endogenous IL-10-producing regulatory B cells develop exacerbated disease and prevent with an increased frequency of Th1/Th17 but a decrease in regulatory T cells

    J. Immunol.

    (2011)
  • M. Davis et al.

    Effect of pemetrexed on innate immune killer cells and adaptive T cells in subjects with adenocarcinoma of the pancreas

    J. Immunother.

    (2012)
  • O. De Henau

    Overcoming resistance to checkpoint blockade therapy by targeting PI3Kγ in myeloid cells

    Nature

    (2016)
  • L. De Monte et al.

    Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer

    J. Exp. Med.

    (2011)
  • D.J. DiLillo et al.

    Maintenance of long-lived plasma cells and serological memory despite mature and memory B cells depletion during CD20 immunotherapy in mice

    J. Immunol.

    (2008)
  • C. Feig

    Targeting CXCL12 from FAP-expressing carcinoma-accociated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer

    Proc. Natl. Acad. Sci. U. S. A.

    (2013)
  • F. Flores-Borja et al.

    CD19+CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation

    Sci. Transl. Med.

    (2013)
  • J.D. Fontenot et al.

    Foxp3 programs the development and function of CD4+CD25+ regulatory T cells

    Nat. Immunol.

    (2003)
  • A. Fukunaqa

    CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma

    Pancreas

    (2004)
  • R.F. Gabitass et al.

    Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13

    Cancer Immunol. Immunother.

    (2011)
  • T.J. Goodwin et al.

    On the article “findings questioning the involvement of Sigma-1 receptor in the uptake of anisamide-decorated particles”

    J. Control. Release

    (2016)
  • C. Guillerey et al.

    NK cells and cancer immunoediting

    Curr. Top. Microbiol. Immunol.

    (2016)
  • A.J. Gunderson

    Bruton tryrosine kinase-dependent immune cell cross-talk drives pancreas cancer

    Cancer Discov.

    (2016)
  • S. Guo et al.

    Co-delivery of cisplatin and rapamycin for enhanced anticancer therapy through synergistic effects and microenvironment modulation

    ACS Nano

    (2014)
  • N. Hiraoka et al.

    Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions

    Clin. Cancer Res.

    (2006)
  • Cited by (0)

    1

    These authors contributed equally.

    2

    Lead Contact.

    View full text