Weekly Digests
‹ Back to February

T cells CRISPRed to perfection for attack on refractory cancer

February 26, 2020

Aiming to improve the safety and efficacy of engineered T cells for the treatment of human cancers, Stadtmauer and Fraietta et al. utilized CRISPR-Cas9 genome editing to knock out PD-1 (PDCD1) and the endogenous TCR α and β chains (TRAC and TRBC) in autologous T cells that also expressed an exogenous TCR targeting the cancer-specific antigen NY-ESO-1. The engineered T cells were tested in a first-in-human clinical trial, and the results were recently published in Science.

Patient T cells were harvested and modified by Cas9/guide RNA complexes (delivered by electroporation) and an HLA-A2*0201-restricted TCR specific for the SLLMWITQC peptide in NY-ESO-1 and LAGE-1 cancer-testis antigens (delivered by lentiviral transduction). The genetically edited autologous T cells were called “NYCE” (NY-ESO-1 transduced CRISPR 3X edited cells). NYCE cells were successfully created for four out of six enrolled patients. One of those four patients experienced rapid disease progression and became ineligible for infusion. Thus, three patients (two with refractory advanced multiple myeloma, and one with refractory metastatic liposarcoma) were infused with the NYCE product. Prior to infusion, patients were pre-treated with lymphodepleting chemotherapy.

Before administration of the engineered T cells to patients, NYCE cells were analyzed in vitro for editing efficiency and potency. Analysis of the CRISPR-Cas9 editing efficiency showed that most mutations were on-target (albeit with efficiencies of 45% [TRAC], 15% [TRBC], and 20% [PDCD1]), and very few off-target mutations were detected. Chromosomal translocations were observed in all manufactured NYCE products, but they declined in frequency in patients over time, suggesting that the translocations did not provide any cell growth advantage. In a co-culture with HLA-A2+NY-ESO-1+ Nalm-6 leukemia cells, NYCE cells efficiently killed target cells in an antigen-specific manner. Consistent with preclinical experiments, T cells with endogenous TCR knockout were more cytotoxic than T cells that retained their endogenous TCR.

NYCE infusions were well tolerated, and no cytokine release syndrome or other side effects related to infusion were observed in any of the three treated patients. The engineered T cells peaked at high levels shortly after infusion and persisted for a long time in all three patients (from three to nine months post infusion), indicating stable engraftment. The average half-life of NYCE cells was 84 days, which was strikingly higher than the ~1 week half-life of NY-ESO-1 engineered T cells reported in other clinical trials. Biopsies showed that NYCE cells trafficked to the tumor (or in the case of multiple myeloma, the bone marrow) at levels close to those in the blood. Despite successful engraftment of PD-1- NYCE cells, no patients exhibited evidence of autoimmunity. While healthy donors usually develop antibody and T cell responses to Cas9, the three treated patients did not – possibly due to low levels of Cas9 in the infused product, or the patients’ immunodeficiency due to previous treatments/lymphodepletion. Lack of reactivity to Cas9 was consistent with the prolonged persistence of the infused product.

The researchers analyzed the transcriptomic evolution of NYCE cells over time in one patient who showed signs of tumor regression. NYCE cells were recovered from the blood at day 10 and at day 113 (~4 months) after infusion. Single-cell RNA sequencing showed a decline in the frequency of gene-edited T cells over time (regardless of whether the cells had a transduced TCR). Most of the decline occurred in the first 10 days, with the frequency of gene-edited cells holding generally stable between 10 days and 4 months post infusion. Cells in the manufactured product had mutations in one, two, or all three targeted sequences. Approximately 25% of engineered T cells expressing the exogenous NY-ESO-1 TCR also had mutated PDCD1 in the initial infusion product, but this percentage decreased to ~5% by 4 months post infusion, which is consistent with studies that have shown that PD-1- T cells are less capable of establishing memory. In this patient, NY-ESO-1-expressing cells increased the expression of IL7R and TCF7 over time, indicating central memory phenotype. This is in contrast with previous findings that showed that NY-ESO-1-expressing T cells, which had not been gene-edited, tended to develop a terminally differentiated phenotype and exhibited characteristics of exhaustion.

In this pilot study, two of the three treated patients had stable disease, which was the best clinical response. One patient had a significant (~50%) and sustained (for 4 months) decrease in an abdominal tumor mass, but had progression in other lesions. Ultimately, all three patients progressed; one of the patients died of progressive disease, and the other two are receiving other therapies. In the two patients with multiple myeloma, there was a decrease in the target antigens (NY-ESO-1 and/or LAGE-1), though residual tumor remained, possibly indicating an on-target effect that resulted in tumor editing. Blood samples obtained from patients three to nine months post infusion and expanded in culture in the presence of the NY-ESO-1 peptide demonstrated antigen-specific cytotoxicity in all three patients.

In this first-in-human clinical trial, Stadtmauer and Fraietta et al. demonstrated the initial safety and feasibility of using CRISPR-Cas9 to edit multiple genes in autologous T cells for the treatment of advanced, refractory cancer. Future studies with more patients will be necessary to assess the full safety profile and the efficacy of this approach.

by Anna Scherer

References:

Stadtmauer E.A., Fraietta J.A., Davis M.M., Cohen A.D., Weber K.L., Lancaster E., Mangan P.A., Kulikovskaya I., Gupta M., Chen F., Tian L., Gonzalez V.E., Xu J., Jung I.Y., Melenhorst J.J., Plesa G., Shea J., Matlawski T., Cervini A., Gaymon A.L., Desjardins S., Lamontagne A., Salas-Mckee J., Fesnak A., Siegel D.L., Levine B.L., Jadlowsky J.K., Young R.M., Chew A., Hwang W.T., Hexner E.O., Carreno B.M., Nobles C.L., Bushman F.D., Parker K.R., Qi Y., Satpathy A.T., Chang H.Y., Zhao Y., Lacey S.F., June C.H. CRISPR-engineered T cells in patients with refractory cancer. Science. 2020 Feb 6.

In the Spotlight...

CLINICAL TRIAL: EV-101: A Phase I Study of Single-Agent Enfortumab Vedotin in Patients With Nectin-4-Positive Solid Tumors, Including Metastatic Urothelial Carcinoma

Rosenberg et al. summarized a phase I, dose escalation trial of enfortumab vedotin (EV) – an ADC conjugate (anti-NECTIN-4–MMAE) – in 155 heavily pretreated patients with solid tumors, including mostly metastatic urothelial carcinoma (mUC). The recommended phase II dose was 1.25mg/kg, dosed 3 times over a 28-day cycle. EV was generally well tolerated, resulting in mostly grade 1–2 AEs, particularly skin (Nectin-4 on-target, off-tumor) and neuropathy (tubulin inhibitor MMAE). In 112 mUC patients receiving 1.25mg/kg EV, ORR was 43% and OS was 12.3 months, and was independent of age, prior anti-PD-(L)1 treatment, and liver mets. Phase II and III trials are underway.

Contributed by Katherine Turner

"UniCAR"-modified off-the-shelf NK-92 cells for targeting of GD2-expressing tumour cells

Mitwasi et al. stably transduced NK-92 cells with ‘universal’ (Uni)CARs comprising an scFv specific for (intracellular antigen) peptide E5B9, a CD28 transmembrane, and a CD28-CD3ζ signaling domain, and built soluble E5B9-tagged anti-disialoganglioside (GD2)-scFV and IgG4 targeting molecules (TMs). FACS showed TMs bind to GD2+ neuroblastoma/melanoma cells and to UniCAR-NK cells via E5B9. TM-bridging of GD2+ tumors to UniCAR-NK cells rapidly induced NK cell secretion of IFNγ and enhanced GD2+ tumor cytolysis in vitro (<1 nM EC50). Injecting nude mice s.c. with UniCAR-NK cells and anti-GM2 TMs inhibited GD2+ neuroblastoma growth.

Contributed by Paula Hochman

REVIEW: CD8+ T cell states in human cancer: insights from single-cell analysis

Motivated by the diversity of intratumoral CD8+ T cells observed by single-cell analysis, van der Leun, Thommen, and Schumacher contextualize these findings into heuristic themes. The major CD8+ T cell subsets include naive-like/memory and cytotoxic/effector, which may or may not be tumor-specific, and dysfunctional, likely tumor-specific cells. Dysfunctional cells are transcriptionally and phenotypically heterogeneous along a continuum but are not non-functional, demonstrating potential for proliferation and chemokine secretion. Some dysfunctional subset(s) are likely responsible for efficacy with checkpoint blockade, and may be useful predictive biomarkers.

Contributed by Alex Najibi

Peripheral CD8+ T cell characteristics associated with durable responses to immune checkpoint blockade in patients with metastatic melanoma

Fairfax et al. show that large TCR clone counts (>5% of the total number of clones) in peripheral CD8+ T cells are predictive of positive response to checkpoint therapy in patients with metastatic melanoma. Transcriptomic analysis revealed overexpression of T cell receptor genes in early samples (day 21) from ICB-responding patients, which was associated with higher number of discrete large clones. Patients with a higher number of large clones in circulation demonstrated increased progression-free and overall survival. Single-cell sequencing showed that cells from large clones display cytotoxic gene overexpression and effector memory phenotypes.

Contributed by Shishir Pant

Mannose receptor (CD206) activation in tumor-associated macrophages enhances adaptive and innate antitumor immune responses

Structural homology screening of antimicrobial host defense peptides identified RP-182 – a synthetic 10-mer amphipathic analog that bound the mannose receptor (CD206) on human and murine macrophages. Binding of RP-182 to CD206 on macrophages induced phagocytosis, autophagy, and apoptosis, resulting in M2 macrophage death or reprogramming of M2 macrophages toward an M1-like phenotype. In mouse models of pancreatic cancer, RP-182 prolonged survival and suppressed tumor growth (dependent on CD8+ T cells), decreased Tregs, increased intratumoral CD8+ T cells, and synergized with gemcitabine to further increase survival.

Contributed by Anna Scherer

Bovine papillomavirus prostate cancer antigen virus-like particle vaccines are efficacious in advanced cancers in the TRAMP mouse spontaneous prostate cancer model

Simons et al. designed a virus-like particle vaccine comprising bovine papillomavirus L1 proteins with docking sites loaded with prostate cancer-associated (PSCA, PAP-1, PAP-2) and neoantigen (SPAS-1) peptides. In TRAMP mice with advanced prostate cancer, the vaccine induced antigen-specific responses, and significantly reduced total prostate weight, both alone and in combination with anti-PD-1; anti-PD-1 alone was ineffective. All treatments increased CD3+ and CD8+ T cell infiltration into tumors, with combination therapy being most effective. Prostate weight negatively correlated with CD8+ T cell numbers in tumors of mice treated with vaccine alone.

Contributed by Anna Scherer

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.