Weekly Digests
‹ Back to May

Targeting NRP2-VEGF to overcome immunosuppression and chemotherapy resistance in aggressive cancers

May 24, 2023

Neuropilin-2 (NRP2) is a transmembrane protein that binds ligands such as vascular endothelial growth factor (VEGF). It is expressed in several tumor types, and overexpressed in aggressive, treatment-resistant tumors, such as triple-negative breast cancer (TNBC), metastatic castration-resistant prostate cancer (mCRPC), and neuroendocrine prostate cancer (NEPC). Two recent papers from the same laboratory, published in Science Translational Medicine, described results on VEGF-specific blockade of NRP2 in these aggressive tumor types to improve immune responses and the efficacy of chemotherapy.

Prostate cancer cells can evade immune destruction by expressing PD-L1, which is seen particularly in more aggressive prostate cancers, and is correlated with poor prognosis and resistance to immune checkpoint blockade (ICB) therapy. Furthermore, these tumors express high levels of NRP2, which is also an independent prognostic factor for worse outcomes.

Wang et al. sorted the metastatic prostate cancer cell line PC3 based on NRP2hi and NRP2lo populations and subjected them to RNAseq. This revealed that gene networks related to the immune response were enriched in NRP2hi cells, including IFNγ signaling and CD274 mRNA (encoding PD-L1). In published human prostate cancer datasets, a positive correlation was detected between NRP2 and CD274 expression. To assess whether this correlation was causal, NRP2-deficient cells were generated using CRISPR-Cas9 or shRNA, which resulted in the downregulation of PD-L1 expression.

The researchers then investigated the immunologic effects of NRP2 expression. Coculture experiments were conducted with human peripheral blood mononuclear cells (PBMCs) stimulated with anti-CD3/CD28 and irradiated PC3 cells with or without NRP2 expression. In these cultures, the proliferation of CD8+ and CD4+ T cells was inhibited when control PC3 cells were present, while NRP3ko PC3 partially restored the proliferation of T cells and IFNγ production. When NRP2 or PD-L1 expression was restored in the KO cells, T cell proliferation was reduced, suggesting the suppression of T cell activation by NRP2 is mediated by PD-L1 expression.

To investigate a correlation between VEGF/NRP2 and PD-L1 expression in aggressive human prostate cancers, published datasets in cBioPortal were analyzed. Levels of NRP2, VEGFA, and VEGFC genes were increased in the aggressive prostate cancer subtypes mCRPC and NEPC. There was also a positive correlation between NRP2 and CD274 expression in metastatic prostate cancer samples. The NRP2hiPD-L1hi cell subsets in patients with metastatic cancer had lower androgen receptor expression and higher NEPC scores than NRP2loPD-L1lo cells, suggestive of a more aggressive phenotype.

Therapeutic targeting of NRP2 is challenging, as it functions as a receptor for multiple ligands, and only the interaction with VEGF has been associated with aggressive tumoral behavior. Therefore, Xu, Lal Goel, Burkart, et al. produced a highly specific monoclonal antibody (aNRP2-10) with a high affinity for the b1 domain of NRP2, which blocks the binding of VEGF-A and VEGF-C to NRP2, but does not block binding of semaphorin, another important NRP2 ligand.

Breast cancer stem cells (CSCs) express high levels of NRP2. The researchers sorted the BT-549 TNBC cell line into NRP2hi and NRP2lo populations. The NRP2hi cells had higher self-renewal potential than the NRP2lo cells. In vitro treatment with aNRP2-10 could significantly inhibit sphere formation of various cell lines and the self-renewal potential of CD44hiCD24lo CSC populations isolated from patient TNBC samples. To assess whether this NRP2 blockade could lower the frequency of CSCs, a transplantation experiment was performed using the murine 4T1 TNBC model. Treatment with aNRP2-28, a mouse-specific surrogate of aNRP2-10, reduced the frequency of CSCs, which was associated with a reduced expression of ZEB1, a CSC marker and regulator of epithelial-to-mesenchymal transition (EMT).

Given its effects on CSCs, which are largely chemoresistant, the researchers assessed whether aNRP2-10 could sensitize TNBC to chemotherapy treatment. aNRP2-10 treatment of 3D MDA-MB-231 cell cultures increased the sensitivity of tumor cells to cisplatin-induced killing. This was further analyzed with other cell lines, but only some of the cell lines “responded” to therapy. The responder cell lines were characterized by higher EMT scores and expression of ZEB1 and NRP2. Likewise, aNRP2-10 could sensitize TNBC patient-derived organoids to 5-fluorouracil (5-FU).

Similar increases in chemosensitization were achieved when NRP2 was downregulated using small interfering RNAs (siRNAs) in TNBC organoids, suggesting that VEGF/NRP2 signaling is a major driver of chemoresistance in these organoids. Gene expression analyses of the TNBC organoids showed that CSC and EMT markers, including ZEB1, were significantly reduced following aNRP-10 treatment, while they increased after 5-FU monotherapy.

In vivo studies with a TNBC xenograft further validated the efficacy of aNRP2-10. Antibody treatment alone did not affect tumor growth, but when combined with cisplatin, tumor growth was reduced. Gene expression analysis of the xenografts showed that CSC and EMT markers were downregulated by aNRP2-10 treatment. These effects were confirmed in another TNBC xenograft model and a patient-derived xenograft (PDX) model in combination with sub-optimal 5-FU treatment. In humanized mice with a TNBC xenograft, aNRP2-10 treatment alone reduced lung metastases.

The aNRP2 antibody was also tested by Wang et al. in comparison to anti-PD-L1 in the transgenic adenocarcinoma mouse prostate (TRAMP) model, which is resistant to ICB treatment. Established tumors were treated with aNRP2-28 or anti-PD-L1. Treatment with anti-PD-L1 did not impact tumor volume, while aNRP2-28 alone reduced tumor burden. This effect was not detected in immunodeficient NSG mice. Tumor histology analyses revealed that aNRP2-28 induced necrosis in tumors, while anti-PD-L1 did not. In the non-necrotic areas of the tumor, treatment with aNRP2-28 reduced PD-L1 expression, and there was an increase in tumor-infiltrating lymphocytes (TIL), including activated effector T cells, suggesting the tumor immune environment turned from “cold” to “hot”.

The therapeutic effects of aNRP2-10 were also assessed in the human prostate cancer setting, using human NEPC patient-derived organoid models. In this model, treatment reduced PD-L1 expression, and increases in tumor cell killing were observed when tumor organoids were cocultured with activated PBMCs.

Finally, a toxicity study performed by Xu, Lal Goel, Burkart, et al. in cynomolgus monkeys showed that aNRP2-10 was well tolerated, with no observed adverse effects at the highest dose tested.

In conclusion, these two studies showed the benefits of specifically targeting NRP2 to block VEGF signaling in aggressive cancer types, inducing antitumor immune responses in cold tumors, and targeting CSCs in breast cancer to improve the efficacy of chemotherapy. These results set the stage for testing various combination strategies with this NRP2/VEGF-targeting antibody in the clinical setting.

Write-up by Maartje Wouters, image by Lauren Hitchings.

References:

Wang M, Wisniewski CA, Xiong C, Chhoy P, Goel HL, Kumar A, Zhu LJ, Li R, St Louis PA, Ferreira LM, Pakula H, Xu Z, Loda M, Jiang Z, Brehm MA, Mercurio AM. Therapeutic blocking of VEGF binding to neuropilin-2 diminishes PD-L1 expression to activate antitumor immunity in prostate cancer. Sci Transl Med. 2023 May 3. 

Xu Z, Goel HL, Burkart C, Burman L, Chong YE, Barber AG, Geng Y, Zhai L, Wang M, Kumar A, Menefee A, Polizzi C, Eide L, Rauch K, Rahman J, Hamel K, Fogassy Z, Klopp-Savino S, Paz S, Zhang M, Cubitt A, Nangle LA, Mercurio AM. Inhibition of VEGF binding to neuropilin-2 enhances chemosensitivity and inhibits metastasis in triple-negative breast cancer. Sci Transl Med. 2023 May 3.

In the Spotlight...

A rationally designed ICAM1 antibody drug conjugate eradicates late-stage and refractory triple-negative breast tumors in vivo

ICAM1 is highly overexpressed in triple-negative breast cancer (TNBC) cells and facilitates receptor-mediated antibody internalization. Guo et al. developed a rationally designed panel of four ICAM1 antibody–drug conjugates using different chemical linkers and warheads for the treatment of TNBC. IC1–MMAE, which has a protease-cleavable linker and a microtubule inhibitor warhead, showed the highest and most consistent efficacy for complete and durable tumor regression and eradication in a series of standard, late-stage, and refractory TNBC models in vivo. IC1–MMAE did not show liver or renal toxicities in systemic treatment at dosages of 1 to 10 mg/kg.

Contributed by Shishir Pant

PSGL-1 attenuates early TCR signaling to suppress CD8+ T cell progenitor differentiation and elicit terminal CD8+ T cell exhaustion

Investigating PSGL-1’s role in regulating CD8+ T cell responses, Hope et al. showed that PSGL-1 co-ligated with the TCR to inhibit proximal TCR signaling via Zap70 to restrain mouse and human CD8+ T cell activation, driving terminal T cell exhaustion. In PSGL-1-deficient conditions compared to WT, CD8+ TILs responded better to low affinity TCR ligands, exhibited enhanced glycolysis, and increased numbers of stem cell-like progenitors with Teff function. Therapeutic PSGL-1 blockade decreased T cell exhaustion and inhibited growth of PD-1 blockade-resistant melanoma suggesting PSGL-1 could be targeted to treat PD-1-non-responsive tumors.

Contributed by Katherine Turner

A first-in-human study of the fibroblast activation protein-targeted, 4-1BB agonist RO7122290 in patients with advanced solid tumors

Melero and Tanos et al. report phase 1 results for RO7122290, a bispecific fusion protein carrying a 4-1BB (CD137) ligand and a fibroblast activation protein α binding site that co-stimulates T cells as a single agent or in combination with atezolizumab in patients with advanced or metastatic solid tumors. RO7122290 demonstrated an acceptable safety profile and the postulated mechanism of action. Treatment-induced increases in intratumoral and peripheral proliferating and activated T cells were accompanied by associated gene expression changes in both arms. Clinical activity was most strongly observed in combination with atezolizumab (2 CRs and 9 PRs), mostly in CPI naive patients.

Contributed by Shishir Pant

NL-201 upregulates MHC-I expression and intratumoral TCR diversity, and demonstrates robust antitumor activity as monotherapy and in combination with PD-1 blockade

NL-201 is a stable IL-2/IL-15 mimetic that does not bind the high-affinity α receptor subunit, to prevent activation of Tregs, and is pegylated to improve pharmacokinetic half-life. Mortales et al. showed that NL-201 induced antitumor immunity in immune-cold mouse tumors. This effect was associated with increased tumoral MHC-I and PD-L1 expression, which depended on effector cell IFNγ production. Combination treatment with anti-PD-1 in several cold murine tumor models revealed synergistic antitumor efficacy. Treatment increased the clonal diversity of tumoral T cells and increased the number of IFNγ-producing CD8+ and CD4+ Th1 cells, but not Tregs.

Contributed by Maartje Wouters

cis-B7:CD28 interactions at invaginated synaptic membranes provide CD28 co-stimulation and promote CD8+ T cell function and anti-tumor immunity

While B7 (CD80/CD86) on APCs is known to engage CD28 on T cells in head-to-head interactions in trans, Zhao et al. found that B7 on T cells similarly engaged CD28 in cis due to endocytosis-related proximity in the invaginating cell membrane. cis-B7:CD28 interactions released CD28 from membrane sequestration and activated CD28 signaling through PKCθ and PI3K binding. PI3K, which was not required for interactions in trans, recruited SNX9 and drove further CD28 endocytosis, thus promoting a positive feedback loop. cis-B7:CD28 signaling promoted cell-autonomous T cell survival, migration, and cytokine production, and contributed to T cell-mediated antitumor efficacy.

Contributed by Lauren Hitchings

OX40L-expressing recombinant modified vaccinia virus Ankara induces potent antitumor immunity via reprogramming Tregs

To induce local antitumor immunity and deplete tumor-infiltrating Tregs, Yang and Wang et al. engineered the modified vaccinia virus Ankara (MVA) with a deletion of the vaccinia E5R gene (an inhibitor of the cGAS/STING pathway) and insertion of transgenes for surface-bound Flt3L and OX40L expression (rMVA). Intratumoral delivery of rMVA in mouse models delayed tumor growth and increased survival. Therapy induced CD8+ and CD4+ T cell activation and reduced the highly suppressive tumoral OX40hi Treg subset. Efficacy was T cell-dependent and improved with anti-PD-L1 cotreatment. A human version (rhMVA) activated T cells and reduced Tregs ex vivo in human Paget’s disease skin biopsies.

Contributed by Maartje Wouters

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.