Weekly Digests
‹ Back to March

An “IF/THEN” CAR T cell that targets only the tumor

March 27, 2019

CD19-targeted CAR T cells can be incredibly effective against B cell malignancies, however, they do not discriminate between malignant and healthy cells. In the case of lineage-restricted CD19, the killing of on-target, off-tumor cells is not a major problem because patients can survive without B cells. A major challenge in treating solid tumor types, however, has been finding tumor antigens that can be targeted without leading to the unintended destruction of essential healthy cells. ROR1 is a promising tumor target in many epithelial malignancies, but its low-level expression on some normal tissues may soon present a toxicity challenge as ongoing clinical trials aimed at this target antigen progress. To get ahead of this potential problem, Srivastava et al. used a mouse model to understand the basis of potential off-tumor toxicity, which led to the design of logic-gated ROR1-targeted CAR T cell system that more selectively target tumor cells while sparing healthy tissues.

To begin, Srivastava et al. treated healthy mice with ROR1-targeted CAR T cells. CAR T cells alone did not induce toxicity, but mice that had been pre-treated with lymphodepleting radiation or chemotherapy died of CAR T cell-mediated toxicity within two weeks following weight loss, a rapid decline in red blood cell and platelet counts, necrosis in the spleen, and damage to bone marrow. Toxicity was found to be dependent on reaching a threshold of functional CAR T cells in vivo, which was dependent on both the extent of lymphodepletion and the initial cell dose.

Using various mouse models, including Rag2-/- mice, ROR1 knockouts, and bone marrow chimeras, Srivastava et al. determined that necrosis in the spleen could be attributed to CAR T cell targeting of ROR1+ stem cell factor (Scf)-producing splenic stromal cells in the context of lymphodepletion (as it occurred in both pre-conditioned wild-type mice and Rag2-/- mice), but that this effect alone did not cause mortality. Mortality only occurred when there was bone marrow failure due to CAR T cell-mediated killing of ROR1+ bone marrow stromal cells, which occurred in the context of cytotoxic pretreatment, but not in Rag2-/- mice. The researchers found that exposure to radiation induced significant upregulation of ROR1 on mesenchymal stem cells, which could explain why bone marrow damage, and the consequent lethal lack of hematopoietic recovery, was prominent only after cytotoxic pre-treatment in vivo.

Having established an understanding of the source of the lethal toxicity of ROR1-targeted CAR T cells in mice, the researchers turned their attention towards finding a solution. To this end, they engineered T cells with a EpCAM-targeted synthetic Notch receptor circuit, which, when activated, drives transcription of the ROR1 CAR. EpCAM is not expressed on ROR1+ bone marrow or spleen stromal cells, but is co-expressed on 4T1 mammary carcinoma transduced to express ROR1, meaning that these “AND” (or more aptly described as “IF/THEN”) gated cells, should be activated by the presence of EpCAM on the tumor, upregulate the ROR1-CAR, and specifically target local tumor cells.

Specific targeting of dual-target-expressing tumor cells by synNotch T cells was evident in vitro. In in vivo studies, ROR1 CAR T cells controlled tumors, but also quickly accumulated in the spleen and bone marrow, inducing lethal toxicity. SynNotch T cells on the other hand, mediated tumor control without signs of tumor necrosis, and mice were able to recover weight, red blood cells, and platelets, indicating that ROR1+ stromal cells in the bone marrow remained functional. Overall, synNotch T cells mediated increased survival over untransduced control T cells or ROR1 CAR T cells, although all mice still ultimately died due to tumor outgrowth. Further, recognizing that EpCAM may not be an ideal target in a clinical setting, as it can be co-expressed with ROR1 on some normal human epithelial tissues, the researchers identified B7-H3, frequently coexpressed with ROR1 on human breast, lung, and ovarian tumors, as a possible alternative target for the synNotch receptor. B7-H3-targeted synNotch T cells exhibited in vitro and in vivo behaviors that were comparable to those of EpCAM-targeted synNotch T cells.

While the synNotch receptor system may limit the off-tumor toxicity of ROR1-targeting CARs, the potential for toxicity remains when malignant and healthy ROR1+ cells are in close proximity. To test the safety of synNotch T cells in such a context, the researchers inoculated mice with an aggressive ROR1+ lymphoma, which metastasizes to the bone marrow. Mice were then treated with CD19-directed synNotch T cells, which, upon target recognition, would trigger expression of the ROR1 CAR. In this model CD19-directed synNotch T cells and more traditional ROR1 CAR T cells exhibited similar antitumor efficacy and lethal toxicity, highlighting the need for spatial segregation of target cells for the synNotch system to alleviate the toxic effects of the ROR1 CAR.

Overall, Srivastava et al. gained a better understanding of potentially lethal toxicity of ROR1-targeted CAR T cells, and designed synNotch T cells that may limit on-target, off-tumor toxicity by more strictly defining tumor cell targets. The synNotch strategy could likely be applied across various cancers and tumor-associated antigens, though it may be most effective in solid, low-burden tumors that have not metastasized, as efficacy requires the spatial segregation of target antigen-expressing tumor and healthy cells.

by Lauren Hitchings

References:

Srivastava S., Salter A.I., Liggitt D., Yechan-Gunja S., Sarvothama M., Cooper K., Smythe K.S., Dudakov J.A., Pierce R.H., Rader C., Riddell S.R. Logic-Gated ROR1 Chimeric Antigen Receptor Expression Rescues T Cell-Mediated Toxicity to Normal Tissues and Enables Selective Tumor Targeting. Cancer Cell. 2019 Mar 18.

In the Spotlight...

A transcriptionally distinct CXCL13+CD103+CD8+ T-cell population is associated with B-cell recruitment and neoantigen load in human cancer

Analyzing ovarian tumor mRNA, Workel and Lubbers et al. identified a distinct CD103+CD8+ T cell population with an activated, exhausted phenotype and high CXCL13 expression. Peripheral blood CD8+ T cells activated in the presence of TGFβ expressed CD103 and secreted CXCL13. In TCGA, higher predicted neoantigen load and CD103+ infiltrate in uterine cancers correlated with B cell and tertiary lymphoid structure (TLS)-associated genes, and CXCL13+CD103+ T cell and TLS genes correlated across various cancers. CXCL13+CD103+ T cells associated with improved survival even in endometrial tumors with low predicted neoantigen load.

Contributed by Alex Najibi

CD40 Ligand-Modified Chimeric Antigen Receptor T Cells Enhance Antitumor Function by Eliciting an Endogenous Antitumor Response

Anti-CD19 CAR T cells engineered to constitutively express CD40L delayed tumor onset and increased survival in immunocompetent mice with leukemia or lymphoma compared with second-generation CAR T cells. CD40L+ CAR T cells recruited and licensed CD40+ APCs in spleens and lymph nodes, leading to increased tumor infiltration and activation of endogenous CD4+ and CD8+ T cells, increased cytokine production, and a sustained immune response that conferred long-term protection against CD19- tumor challenge. CD40L+ CAR T cells did not require preconditioning and no increase in toxicity was observed.

Enhancing the antitumor functions of invariant natural killer T cells using a soluble CD1d-CD19 fusion protein

Invariant natural killer T cells (iNKT) are able to recognize and kill cells presenting the lipid agonist αGC on CD1d. To exploit iNKT cells in a CD1d-independent manner, Das et al. developed a soluble bispecific fusion protein linking the CD1d molecule to a CD19 scFv. When loaded with αGC, the fusion protein engaged iNKT cells in vitro, inducing activation, cytokine production, proliferation, and specific killing of CD19+CD1d- target cells, including EBV-immortalized B cells. Activated iNKT cells promoted DC maturation, driving the activation of a broader immune response. In vivo, the fusion protein controlled CD19+CD1d- tumor growth.

New epitopes in ovalbumin provide insights for cancer neoepitopes

Using ovalbumin (OVA) as a canonical neoantigen of the mouse, Karandikar et al. studied in vivo the immunogenicity and physiological presentation of predicted epitopes as epitope-length peptides or in the context of whole OVA, and identified several novel epitopes. Mice bearing progressing OVA-expressing E.G7 tumors showed no CD8+ responses to any OVA epitopes, including SIINFEKL, unless Tregs were depleted with an anti-CTLA-4 antibody. Prophylactic immunization with OVA or SIINFEKL, but not any other identified epitope, protected against E.G7 tumors, however, therapeutic vaccination with SIINFEKL did not reduce tumor growth.

An antibody-drug conjugate directed to the ALK receptor demonstrates efficacy in preclinical models of neuroblastoma

Finding that wild-type or mutated anaplastic lymphoma kinase (ALK) is expressed on the surface of most neuroblastoma cells, but not on normal tissue, Sano et al. developed an antibody-drug conjugate (CDX-0125-TEI) consisting of an anti-ALK mAb coupled via a cleavable linker to a thienoindole (a DNA alkylating agent). In vitro, CDX-0125-TEI killed ALK+ human neuroblastoma-derived cells by inducing DNA damage and apoptosis. In mice with patient-derived xenografts, CDX-0125-TEI was well tolerated and delayed tumor growth independent of ALK mutation status, although all tumors progressed.

REVIEW: Host tissue determinants of tumour immunity

Salmon et al. thoroughly review factors in tissue-specific microenvironments that may influence tumor development and immune response across disease sites. These factors include the composition and functional state of tissue-resident non-immune cells (such as cancer-associated fibroblasts) and immune cells (such as macrophages); tumor-infiltrating immune cells; tertiary lymphoid structures; vascular features; nerve cell infiltration; tissue-specific antigenic profile; and the local microbiome. These factors can dramatically impact patient survival and must be considered when designing immunotherapeutic strategies for cancer.

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.