Skip to main content

Advancing Humanized NSG Mice for Immuno-Oncology Efficacy Testing

HuMice

Assessing novel therapeutics that function to modulate the interaction between the human immune system and cancer cells requires advanced preclinical efficacy testing platforms. Immunologically humanized NSGTM and its variant strains are a powerful system that facilitate cutting-edge in vivo preclinical testing of a wide range of immuno-oncology therapeutic candidates.

Taking advantage of NSGTM and newly available variant strains engrafted with human immune cells will enable you to design highly targeted and human biology specific experiments. JAX scientific experts can assist in facilitating selection of the most appropriate platform for your needs.

Oncology therapeutics designed to activate the immune system against tumors are a promising strategy for cancer treatment. To advance these novel therapeutics, specialized preclinical models are crucial in testing efficacy and providing translationally-relevant data. For example, co-engrafting human tumors with human immune cells into the NSGTM (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ,  JAX Stock# 005557) and its variant strains creates a powerful system to interrogate the immunomodulatory effects of human-specific experimental treatments in a preclinical setting.

JAX researchers recently demonstrated this kind of treatment with the PD-1 inhibitor pembrolizumab. They were able to suppress tumor growth in lung, sarcoma, bladder, breast cancer PDX, and a breast cancer cell line when co-engrafted in CD34+ hematopoietic stem cell (HSC) humanized NSGTM mice (Wang et al., 2018). The response to pembrolizumab was variable by HSC donor, highlighting the importance of using multiple HSC donors across all arms in such studies (Wang et al., 2018). In another experiment, treatment with an antibody directed against the costimulatory molecule GITR in CD34+ HSC-engrafted NSGTM mice co-engrafted with the human melanoma cell line SK-MEL-5 suppressed tumor growth and reduced intratumoral T regulatory cell number concurrent with increased activation of CD8+ T cells, mirroring what had been observed in syngeneic model systems (Mahne et al., 2017).

Humanized NSGTM have also proven to be a valuable platform for assessment of anti-tumor T cell activity using both chimeric antigen receptor T cells (CAR-T) and bi-specific T cell engagers (BiTE). In the study by Jin et al., CD70 CAR-T cells were modified to express receptors for IL-8 to enhance intratumoral T cell migration as expression of this chemokine is increased in tumor tissue and can be stimulated by radiation. These CAR-T cells were administered to NRG (NOD.Cg-Rag1tm1Mom Il2rgtm1Wjl/SzJ  JAX Stock# 007799) or NSG-B2m (NOD.Cg-B2mtm1Unc Prkdcscid Il2rgtm1Wjl/SzJ, JAX Stock# 010636) carrying U87 glioma cells, PANC-1 pancreatic tumor cells, or SK-OV-3 ovarian tumor cells after local radiation treatment (Jin et al., 2019). Intratumoral migration of CAR-T was increased and tumor burden was diminished in treated mice (Jin et al., 2019).

In another study, efficacy of a BiTE was assessed in NSGTM mice engrafted with hematological cancers known to express the intracellular tumor marker WT1, where an epitope of the tumor marker is expressed on the cell surface in the context of the MHC I molecule HLA-A2 (Dao et al., 2017). Dao and colleagues demonstrated efficacy of a BiTE directed against WT1 displayed by the tumor and CD3 on human T cells. Interestingly, the authors developed and used an Epstein Barr Virus-specific human T cell population to prevent complications with Graft Versus Host Disease (Dao et al., 2015).

JAX now offers a simplified approach to avoiding acute GvHD when using human PBMC’s in these types of studies.  A new NSGTM variant strain harboring null mutations in mouse MHC I and MHC II (NOD.Cg-Prkdcscid H2-Ab1em1Mvw H2-K1tm1Bpe H2-D1tm1Bpe Il2rgtm1Wjl/SzJ, JAX Stock# 025216) has significantly delayed and diminished GvHD following human PBMC engraftment (Brehm et al., 2019). This platform offers a significant advantage in immuno-oncology studies requiring mature human T cells by enabling much longer study duration and fewer study complications. This model system can be leveraged for preclinical CAR-T efficacy studies by facilitating longer study duration and clearer results.

The Jackson Laboratory In Vivo Services offers oncology efficacy testing services using CD34+ HSC and PBMC humanized NSGTM and NSGTM variant strains, in addition to providing study-ready humanized mice to propel your research and discovery. Reach out to our team to discuss your preclinical testing needs for your CAR-T and BiTE candidates!

REFERENCES

Brehm, M.A., Kenney, L.L., Wiles, M.V., Low, B.E., Tisch, R.M., Burzenski, L., Mueller, C., Greiner, D.L., Shultz, L.D., 2019. Lack of acute xenogeneic graft- versus-host disease, but retention of T-cell function following engraftment of human peripheral blood mononuclear cells in NSG mice deficient in MHC class I and II expression. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 33, 3137–3151. https://doi.org/10.1096/fj.201800636R

Dao, T., Korontsvit, T., Zakhaleva, V., Jarvis, C., Mondello, P., Oh, C., Scheinberg, D.A., 2017. An immunogenic WT1-derived peptide that induces T cell response in the context of HLA-A*02:01 and HLA-A*24:02 molecules. Oncoimmunology 6, e1252895. https://doi.org/10.1080/2162402X.2016.1252895

Dao, T., Pankov, D., Scott, A., Korontsvit, T., Zakhaleva, V., Xu, Y., Xiang, J., Yan, S., de Morais Guerreiro, M.D., Veomett, N., Dubrovsky, L., Curcio, M., Doubrovina, E., Ponomarev, V., Liu, C., O’Reilly, R.J., Scheinberg, D.A., 2015. Therapeutic bispecific T-cell engager antibody targeting the intracellular oncoprotein WT1. Nat. Biotechnol. 33, 1079–1086. https://doi.org/10.1038/nbt.3349

Jin, L., Tao, H., Karachi, A., Long, Y., Hou, A.Y., Na, M., Dyson, K.A., Grippin, A.J., Deleyrolle, L.P., Zhang, W., Rajon, D.A., Wang, Q.J., Yang, J.C., Kresak, J.L., Sayour, E.J., Rahman, M., Bova, F.J., Lin, Z., Mitchell, D.A., Huang, J., 2019. CXCR1- or CXCR2-modified CAR T cells co-opt IL-8 for maximal antitumor efficacy in solid tumors. Nat. Commun. 10, 4016. https://doi.org/10.1038/s41467-019-11869-4

Mahne, A.E., Mauze, S., Joyce-Shaikh, B., Xia, J., Bowman, E.P., Beebe, A.M., Cua, D.J., Jain, R., 2017. Dual Roles for Regulatory T-cell Depletion and Costimulatory Signaling in Agonistic GITR Targeting for Tumor Immunotherapy. Cancer Res. 77, 1108–1118. https://doi.org/10.1158/0008-5472.CAN-16-0797

Wang, M., Yao, L.-C., Cheng, M., Cai, D., Martinek, J., Pan, C.-X., Shi, W., Ma, A.-H., De Vere White, R.W., Airhart, S., Liu, E.T., Banchereau, J., Brehm, M.A., Greiner, D.L., Shultz, L.D., Palucka, K., Keck, J.G., 2018. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 32, 1537–1549. https://doi.org/10.1096/fj.201700740R