Weekly Digests
‹ Back to April

NKp46 finds its ligand in ecto-CRT on stressed and senescent cells

April 19, 2023

Natural killer (NK) cells are known for their ability to recognize and kill targets that are transformed, infected, stressed out, or senescent, and while NK cells are known to act through several NK cell receptors, many of the ligands that activate those receptors have remained elusive. In a recent study published in Nature, Sen Santara and Lee et al. identified ecto-calreticulin (CRT), a product of ER stress, as an activating ligand of NKp46 that contributes to NK cell-mediated killing of stressed and senescent cells.

To begin, Sen Santara and Lee et al. cultured human peripheral blood NK cells with human JEG-3 choriocarcinoma cells. When JEG-3 cells were infected with ZIKV, which replicates in the ER and causes ER stress, NK cell degranulation and JEG-3 killing were enhanced. This effect was abrogated with the addition of ER stress inhibitors, and was not observed with infections that did not induce ER stress. To identify the NK cell receptor(s) that recognized ER stress, NKp46, NKp30, NKp80, NKG2D, DNAM-1, and 2B4 were each blocked, but only the blockade of NKp46 strongly inhibited NK cell killing of ER-stressed target cells. This effect was confirmed using NCR1 (encoding NKp46) knockout models, and with specific binding of NKp46–Ig (NKp46 fused to the Fc region of human IgG1) to ZIKV-infected cells.

Next, the researchers used NKp46–Ig and NKG2D–Ig to pull down cross-linked ligands from ZIKV-infected JEG-3 cell membranes, and found that the NKp46–Ig alone pulled down a band containing calreticulin (CRT) and protein disulfide isomerases (PDI; PDIA1, and PDIA3), which are known to selectively translocate and associate on the cell surface during ER stress. Increased expression of CRT and PDI was confirmed in JEG-3 cells infected with ZIKV or treated with tunicamycin (an ER stressor). Increased NKp46–Ig binding to ecto-CRT was also observed in JEG-3 cells treated with oxaliplatin (an immunogenic cell death [ICD]-inducing chemotherapy). Importantly, the researchers also showed that binding between ecto-CRT on oxaliplatin-treated cells and NKp46 on NK cells induced downstream NKp46 signaling – an effect that was lost with NKp46 knockout or CRT blockade – and mediating NK cell killing.

Digging deeper into this interaction, the researchers evaluated the three domains that make up CRT (N, P, and C), and showed that NKp46 binds specifically to the P domain. Modeling of CRT binding to NKp46 suggested that the P domain, which forms a hairpin structure, may insert itself into a hinge region between the two immunoglobulins that make up NKp46, forming salt bridges. Using cells expressing mutant forms of CRT, the researchers specifically identified the residues D237 and D258 within the P domain as being essential to NKp46–ecto-CRT binding and subsequent effects.

Parsing out the roles of CRT and PDI, the researchers used small interfering RNA to knockdown CALR and PDIA3. While CALR knockdown reduced ecto-CRT on ZIKV-infected JEG-3 cells by about 65%, PDIA3 knockdown reduced ecto-CRT by only 20%. Further, only CALR knockdown reduced NKp46–Ig binding to and killing of ZIKV-infected cells. Similar effects were observed using blocking antibodies.

To determine whether their findings in human cells could be replicated in murine cells, Sen Santara and Lee et al. treated B16F10 melanoma cells (B16) with cisplatin (a non-ICD-inducing drug that does not increase ecto-CRT) or oxaliplatin (an ICD-inducing drug that does increase ecto-CRT). Both cisplatin- and oxaliplatin-treated cells showed increased susceptibility to NK cell-mediated killing. Blocking CRT or NKp46 abrogated this effect only in the oxaliplatin-treated cells, while blocking NKG2D abrogated killing only in the cisplatin-treated cells. Similar effects were observed using NKp46 and NKG2D knockout NK cells, suggesting that in murine NK cells, NKp46 recognition of ecto-CRT contributes to killing of cells treated with an ICD-inducing drug, but not a non-ICD-inducing drug.

Next, the researchers developed a line of engineered B16 cells that stably expressed high levels of the CRT ectodomain with a C-terminal GPI anchor. In line with their high expression of ecto-CRT, B16-GPI-CRT cells showed increased conjugation with NK cells, dependent on NKp46, and showed evidence of capping (cross-linking-induced movement of groups of cross-linked molecules to one end of the cell) of NKp46 and the integrin CD49 on NK cells with CRT on the target. In vitro, B16-GPI-CRT cells also showed slightly increased colony formation, migration, and invasiveness compared to WT B16 cells, suggesting that ecto-CRT may support malignancy.

In vivo, B16-EV (control) and B16-GPI-CRT tumors grew similarly in Ncr1−/− mice, while in WT mice, B16-GPI-CRT tumor growth was suppressed. Numbers of tumor-infiltrating NK cells, CD8+ T cells, and TAMs were similar between tumors, but a higher portion of infiltrating NK cells in the WT tumors expressed perforin, IFNγ, and TNF, suggesting increased target recognition. Similar patterns were observed in mice bearing B16 tumors treated with doxorubicin (ICD-inducing). B16-GPI-CRT tumors also developed fewer metastases than control tumors in WT mice – an effect that was dependent on NK cells and NKp46. Interestingly, B16-GPI-CRT tumors developed more metastases than control tumors in the Ncr1-/- mice, possibly due to the enhanced malignancy-associated properties that these cells demonstrated in vitro. Myeloid cell depletion did not affect metastases in WT mice with either tumor type, but did increase metastases from both tumor types in Ncr1-/- mice, suggesting that NKp46-mediated NK cell control may be the dominant antitumor mechanism, while myeloid cell protection compensates in its absence.

Finally, the researchers investigated the role of NKp46–ecto-CRT binding in NK cell detection and destruction of senescent cells. To this end, trametinib (T, a MEK inhibitor) and palbociclib (P, a CDK4/6 inhibitor) (T + P) were used on A549 human lung cancer cells, inducing senescence, ER stress, and ecto-CRT, along with NKG2D ligands. As in previous models, NK cells showed increased killing of T+P-treated cells compared to untreated cells, and blockade of NKp46, CRT, or NKG2D reduced NK cell-mediated killing, with simultaneous blockade of NKp46 and NKG2D reducing killing to background levels. Similar results were observed in mice bearing KP lung tumors, where T+P suppressed tumor growth, dependent on NKp46. While NKG2D also played a role in controlling these tumors, the role of NKp46 was found to be dominant. Similar patterns were observed with other senescence-inducing drugs, depending on whether they induced ER stress.

Together these results show that NK cells recognize stressed and senescent cells through NKp46 binding to ecto-CRT, which is upregulated during ER stress in both human and murine cells. This fundamental biological mechanism is an important step forward in understanding NK cell biology, and will likely be important in understanding and optimizing numerous cancer immunotherapies involving NK cells, and in developing future immunotherapies that could exploit this pathway.

Write-up and image by Lauren Hitchings

References:

Sen Santara S, Lee DJ, Crespo Â, Hu JJ, Walker C, Ma X, Zhang Y, Chowdhury S, Meza-Sosa KF, Lewandrowski M, Zhang H, Rowe M, McClelland A, Wu H, Junqueira C, Lieberman J. The NK cell receptor NKp46 recognizes ecto-calreticulin on ER-stressed cells. Nature. 2023 Apr.

In the Spotlight...

Computational prediction of MHC anchor locations guides neoantigen identification and prioritization

To improve neoantigen prediction methods, Xia et al. conducted a systematic in silico analysis, buttressed with structural analysis and binding measurements, to identify and score HLA anchor positions for multiple alleles. With the assumption that central tolerance eliminates TCRs identifying the surface of wild-type peptide:HLA complexes, mutated neoantigen peptides with varying positions of the mutation in the peptide were categorized into different scenarios that differentially impacted that surface and would be favored or not to be immunogenic. Using positional information was predicted to significantly re-prioritize neoantigen ranking.

Contributed by Ed Fritsch

An optimized IL-12-Fc expands its therapeutic window, achieving strong activity against mouse tumors at tolerable drug doses

Gutierrez et al. generated a monovalent murine (mDF6006) and human (DF6002) IL-12-Fc fusion protein with abrogated Fcγ receptor binding to further increase the half-life and reduce non-native interactions. mDF6006 exhibited prolonged serum half-life, extended serum IFNγ induction, increased T and NK cell infiltration, decreased immunosuppressive myeloid cells in the tumor, showed potent antitumor activity as a single agent against large ICB-resistant tumors, and sensitized checkpoint-resistant tumors to PD-1 blockade. Fully human DF6002 similarly demonstrated an extended half-life and a protracted IFNγ profile in non-human primates.

Contributed by Shishir Pant

T(STEM)-like CAR-T cells exhibit improved persistence and tumor control compared with conventional CAR-T cells in preclinical models

To improve CAR T cell products currently used in phase I clinical trials for the treatment of solid tumors, Meyran and Zhu et al. developed a protocol to generate TSTEM-like CAR T cells with a distinct memory T cell progenitor gene  expression profile. TSTEM-like CAR T cells demonstrated improved proliferative capacity and cytokine section, even after chronic antigen stimulation in vitro, and increased persistence and tumor control in subcutaneous and orthotopic NSG models, which was enhanced in combination with anti-PD-1. The presence of CD4+ T cells during the CAR T cell production phase was crucial for the generation of fully functional CD8+ TSTEM-like CAR T cells.

Contributed by Ute Burkhardt

Neoadjuvant enoblituzumab in localized prostate cancer: a single-arm, phase 2 trial

Enoblituzumab is a humanized, Fc-engineered, monoclonal antibody targeting B7-H3, which mediates ADCC. Shenderov et al. performed a phase 2 trial assessing enoblituzumab therapy before surgery in 32 previously untreated patients with prostate cancer. Treatment was well tolerated (12% grade 3 adverse events), and treatment did not affect surgery timing or outcomes. The PSA0 rate at 1 year post-surgery was 66%, and 50% of patients had Gleason score downgrade. Tumor and peripheral blood TCR sequencing revealed an association with PSA0 outcomes, and TME analysis revealed immune inflammation upregulation, including increases in CD8+ T cells.

Contributed by Maartje Wouters

Collagen-anchored interleukin-2 and interleukin-12 safely reprogram the tumor microenvironment in canine soft tissue sarcomas

To improve the therapeutic window of IL-2 and IL-12, Stinson and Sheen et al. evaluated canine-ized collagen-binding IL-2 and IL-12 in pet beagles with spontaneous soft tissue sarcomas. Intratumoral cytokines were given at different intervals before tumor excision, and dynamic changes in tumor tissue were assessed by IHC and nanostring RNA. Adverse events were mild (grade 1/2), and transient increases in T and NK cells with cytotoxic immune functions and increases in CPI gene expression (PD-1, CTLA-4 and PD-L1) and IDO (>100x) were observed. Enhanced survival was seen in the B16F10 mouse melanoma model after collagen-IL-2/12 and CPI treatment.

Contributed by Katherine Turner

GD2-CART01 for Relapsed or Refractory High-Risk Neuroblastoma

In a phase 1-2 trial, 27 young patients with r/r neuroblastoma were treated with GD2-targeted, 4-1BB- and CD28-costimulated CAR T cells containing an inducible suicide gene. Treatment was safe; the majority of patients experienced mild cytokine release syndrome, and activation of the suicide gene in one patient depleted CAR T cells within 4h. GD2-CAR T cells expanded in vivo, peaking in blood at 2 weeks, and detectable in blood, bone marrow, and even CSF for months after infusion. 11 patients received multiple CAR T doses with similar safety and expansion profiles. 3-year OS was 40%, with complete response in a third of the patients.

Contributed by Alex Najibi

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.