Weekly Digests
‹ Back to March

Bee venom throws a one-two punch to kill the tumor

March 18, 2020

Targeting the tumor where it lives and turning the tumor against itself was what Yu and Dai et al. had in mind when they developed a one-two punch nanovaccine comprising a lipid nanoparticle (NP) loaded with melittin (a peptide found in European bee venom), called α-melittin-NP. The vaccine, which contained no tumor antigens, promoted tumor cell lysis, releasing whole-cell tumor antigens, and targeted to the lymph nodes, where it stimulated antigen-presenting cells and elicited a systemic antitumor response. The results were recently published in Nature Communications.

Melittin is a small (~2500 dalton) host defense peptide in bee venom that stimulates various aspects of immunity. By itself, melittin has only a narrow safe dose range, with its major side effect being rupture of red blood cells (hemolysis). To overcome this toxicity and improve delivery to lymph nodes, Yu and Dai et al. incorporated melittin in 10-20 nm nanoparticles composed of phospholipids. Imaging data showed that after a subcutaneous injection in mice, α-melittin-NPs and α-peptide-NPs (containing a control peptide) accumulated in inguinal and axillary draining lymph nodes. As expected, free melittin did not target to the lymph nodes, and instead was directly absorbed into the blood, where it led to hemolysis. Within the lymph nodes, α-melittin-NPs and α-peptide-NPs were mostly taken up by macrophages and dendritic cells (DCs), but very few of them ended up in B cells or T cells. Moreover, α-melittin-NPs increased the percentage of activated macrophages and DCs, while free melittin and α-peptide-NPs did not.

Exploring the safety of α-melittin-NPs, the researchers incubated α-melittin-NPs or free melittin with bone marrow-derived DCs (BMDCs), bone marrow-derived macrophages (BMDMs), or B16F10 melanoma cells. They observed that free melittin was cytotoxic to all three types of cells, however, α-melittin-NPs killed the tumor cells while sparing the BMDCs and BMDMs at a certain concentration range. The researchers hypothesized that there may be two reasons for the differential cytotoxicity to these cell populations. First, cancer cells have a slightly higher negative charge in their membranes than normal cells, and therefore may be more likely to attract α-melittin-NPs. Second, Yu and Dai et al. observed that α-melittin-NPs were endocytosed by BMDMs and BMDCs (where they localized in intracellular membranes), but were distributed in the cell membrane of the tumor cells, making the tumor cells more susceptible to cell membrane disruption and necrosis. Using mice inoculated with fluorescently labeled B16F10 tumor cells, the researchers confirmed that both free melittin and α-melittin-NPs made the tumor cell membrane more permeable and induced the release of tumor antigens in vivo.

Next, the researchers turned to mouse models to assess the antitumor effect of α-melittin-NPs in vivo. In a bilateral flank B16F10 tumor model (where the tumor in the right flank was implanted 4 days after the tumor in the left flank), intratumoral injection of α-melittin-NPs into the left flank drastically suppressed tumor growth in both the injected tumor (by 95%) and the distant, non-injected tumor (by 92%) compared with control (PBS injection) at 20 days post left tumor inoculation. A smaller inhibitory effect on tumor growth was observed with free melittin (37% in the injected tumor and 66% in the distant tumor). At 60 days post tumor inoculation, among the mice treated with α-melittin-NPs, 70% were tumor-free in the left flank and 50% were tumor-free in the right flank. In contrast, among mice treated with free melittin, 10% were tumor-free in the left flank and none were tumor-free in the right flank. Upon tumor rechallenge, mice treated with α-melittin-NPs had prolonged survival, and 20% of mice treated with α-melittin-NPs completely rejected the tumors.

Examining the antitumor response in more detail, the researchers observed that α-melittin-NPs were mainly found in the injected tumor site, and were practically undetectable in the contralateral tumor, suggesting that the reduced growth of the distant tumor was due to a systemic immune response, and not due to direct tumor cell killing by α-melittin-NPs. Lymphocytes collected from tumor-draining lymph nodes and restimulated with B16F10 tumor lysate-pulsed DCs showed that α-melittin-NPs increased the frequencies of IFNγ+CD8+ T cells (12-fold) and IFNγ+CD4+ T cells (7-fold) 21 days after tumor inoculation. In contrast, free melittin only moderately increased the frequency of IFNγ+CD8+ T cells (3-fold). Analysis of serum collected at day 21 and incubated with B16F10 tumor cells showed that there was no difference in the increase of IgG+ cells between free melittin and α-melittin-NP treatment groups, indicating that the enhanced antitumor effect observed with α-melittin-NPs was mainly due to cellular immunity and not an antibody response. Furthermore, by inoculating mice with two different types of tumors (E0771 breast cancer cells in the left flank, B16F10 cells in the right flank), Yu and Dai et al. showed that the α-melittin-NP-induced systemic antitumor response was antigen-specific, as the treatment failed to inhibit growth of the distant tumor when the injected and distant tumor types were mismatched.

A more in-depth analysis of leukocytes infiltrating the distant tumor in mice with matching tumors in both flanks revealed that α-melittin-NPs significantly altered the microenvironment of the distant tumor. 14 days after treatment of the left tumor, α-melittin-NPs increased the number of CD8+ T cells, CD4+ T cells, NK cells, and monocytes. Free melittin increased the number of CD4+ T cells and monocytes, but not CD8+ T cells, compared with PBS treatment. The α-melittin-NP-induced intratumoral leukocyte infiltration was driven by increased levels of chemokines (CCL3, CCL4, CXCL5, CXCL10, CCL22) that play a role in the recruitment of T cells, NK cells, and monocytes. The treatment also increased levels of proinflammatory cytokines, including TNFα, IL-1β, IL-1α, and IL-6. These data suggest that α-melittin-NPs led to an inflamed tumor microenvironment in the non-injected tumor.

The results of this study suggest that α-melittin-NPs could serve as an effective in situ nanovaccine without the need for antigen loading for cancer types that are accessible for intratumoral injection, providing the benefits of inducing a broad, systemic immune response, delaying tumor growth, and mediating some complete regressions of uninjected tumors, while requiring a relatively simple preparation and exerting no apparent side effects.

by Anna Scherer

References:

Yu X., Dai Y., Zhao Y., Qi S., Liu L., Lu L., Luo Q., Zhang Z. Melittin-lipid nanoparticles target to lymph nodes and elicit a systemic anti-tumor immune response. Nat Commun. 2020 Feb 28.

In the Spotlight...

Annexin A5 as an immune checkpoint inhibitor and tumor-homing molecule for cancer treatment

Kang and Park et al. treated tumor-bearing mice with cisplatin, tumor-specific-antigen peptides (intratumorally), and Annexin A5 (Anx, a phosphatidylserine ligand). The treatment reversed cisplatin-induced Treg, MDSC, TGFβ, and PD-L1 immunosuppression, and enhanced tumor control and tumor-infiltrating (TI) CD4+ and antigen-specific CD8+ T cells, M1 macrophages, and TNFα levels. Systemic delivery of antigen as a tumor-homing Anx/tumor-antigen fusion protein instead of intratumoral peptides further boosted tumor control as well as systemic and TI tumor-specific immune responses, and synergized with other checkpoint inhibitors.

Contributed by Paula Hochman

NOX4 inhibition potentiates immunotherapy by overcoming cancer-associated fibroblast-mediated CD8 T-cell exclusion from tumours

Ford et al. generated cancer-associated fibroblast (CAF)-rich murine models of lung, breast, and colorectal cancers to show that CAFs promote tumor growth and resistance to immunotherapy (anti-cancer vaccination and anti-PD-1) by suppressing CD8+ T cell infiltration in the tumors. CD8+ T cells from CAF-rich tumors upregulated CTLA-4 and were predominantly localized at tumor margins. NOX4 or TGFβ1 inhibition prevented CAF differentiation, but only NOX4 inhibition normalized CAFs into a quiescent fibroblast-like state, promoted CD8+ T cell infiltration, and re-sensitized CAF-rich tumors to anti-cancer vaccination and anti-PD-1 therapy.

Contributed by Shishir Pant

CTLA-4 blockade boosts the expansion of tumor-reactive CD8+ tumor-infiltrating lymphocytes in ovarian cancer

With the goal of improving adoptive cell therapy (ACT) for ovarian cancer, Friese et al. evaluated the impact of CTLA-4 blockade on yield and potency of tumor-reactive TILs. Adding anti-CTLA-4 to the initial TIL culture of ovarian tumor fragments from 14 patients, and/or during the subsequent rapid expansion phase, resulted in greater numbers of CD8+ TEM cells with increased tumor reactivity, compared to standard TIL culture (IL-2 only). Early CTLA-4 blockade did not alter T cell activation, exhaustion and co-stimulatory markers, or the TCR repertoire, suggesting CTLA-4 blockade during initial TIL expansion could improve ACT.

Contributed by Katherine Turner

Treatment of an Aggressive Orthotopic Murine Glioblastoma Model with Combination Checkpoint Blockade and a Multivalent Neoantigen Vaccine

Comparing murine glioblastoma models, Liu et al. found increased macrophage infiltration, tumor-infiltrating lymphocyte (TIL) dysfunction, and anti-PD-L1 therapy resistance in orthotopic CT2A versus GL261 tumors. Whole-exome and RNA sequencing identified 29 CT2A candidate neoantigens, of which 13 induced CD8+ T cell responses after vaccination, and three generated endogenous responses; stability of the peptide-MHC complex correlated with immunogenicity. Anti-PD-L1 therapy along with vaccination against the three identified peptides boosted neoantigen-specific CD8+ TILs and extended survival of CT2A-bearing mice.

Contributed by Alex Najibi

Intratumoral Delivery of a PD-1-blocking scFv encoded in Oncolytic HSV-1 Promotes Antitumor Immunity and Synergizes with TIGIT Blockade

Based on observations that an oncolytic herpes virus (OVH) induced upregulation of PD-L1 in the TME, Lin and Ren et al. developed an OVH that expressed a single-chain variable fragment against PD-1 (OVH-aMPD-1). In vitro, OVH-aMPD-1 promoted phagocytosis of cancer cells, cross-presentation by DCs, and immunogenic cell death. When delivered intratumorally into mice bearing bilateral tumors, the scFv was expressed, and infiltration of activated CD155(TIGIT)+ MDSCs, CD4+, and CD8+ T cells (including tumor-specific CD8+ T cells) was enhanced, mediating bilateral antitumor activity. OVH-aMPD-1 also synergized with TIGIT blockade.

Contributed by Lauren Hitchings

Melanoma-secreted lysosomes trigger monocyte-derived dendritic cell apoptosis and limit cancer immunotherapy

Investigating why large tumors often fail to respond to immunotherapy, Santana-Magal et al. found that in established murine melanomas, tumor-infiltrating DCs (TIDCs), most of which were monocyte-derived, did not migrate to sentinel lymph nodes. TIDCs were found to be essential for licensing cytotoxic activity by CD8+ TILs, however, TIDCs in large tumors underwent apoptosis over time due to excessive phagocytosis of lysosomes. In ACT studies, agonist anti-CD40 enhanced the numbers and viability of MoDCs, improving antitumor efficacy. Analysis of patient data showed that activated MoDCs in tumors correlated with improved survival.

Contributed by Lauren Hitchings

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.