Weekly Digests
‹ Back to December

MEK inhibition induces stem cell memory in CD8+ T cells

December 9, 2020

In the past, studies have shown that inhibition of MEK (MEKi) increases antitumor immunity and leads to the accumulation of activated CD8+ T cells in the TME. While this strategy is known to synergize with checkpoint inhibitors and adoptive cell transfer, little is known regarding exactly how MEKi affects T cell function, differentiation, or memory generation. In a study recently published in Nature Immunology, Verma et al. investigated the effects of MEKi on T cells and how it contributes to antitumor immunity.

To begin, Verma et al. showed that the addition of MEKi (using the MEK1/2 inhibitor selumetinib) to vaccination against known antigens (gp100 in B16F10 melanoma models and HPV16E7 in TC-1 models) in tumor-bearing mice increased total CD8+ T cells (many of which were antigen-specific and expressed granzyme B), reduced tumor burden, and improved survival. The CD8+ T cell population showed increased markers of activation (CXCR3, IL-2Rβ, OX40) and reduced markers of inhibition (CTLA4, TIM3, LAG3), though differences in PD-1 expression were not observed with the addition of MEKi to vaccine. 

Investigating how MEKi enhances CD8+ T cells, Verma et al. turned to metabolic fitness as a possible explanation. MEKi treatment in mice increased mitochondrial mass in CD8+ T cells from the TME, and ex vivo, MEKi treatment of pMEL-1 cells increased the number of mitochondria per cell. MEKi-treated cells had intact redox machinery, tightly packed cristae in mitochondria, higher oxygen consumption rates, and lower extracellular acidification rates, indicative of enhanced mitochondrial fitness and respiratory capacity. Further analysis of metabolic activity showed that glucose uptake did not change with MEKi treatment; however, fatty acid uptake and fatty acid-mediated metabolism were increased. 

Given that metabolic fitness, respiratory capacity, and fatty acid oxidation have previously been associated with immune memory, Verma et al. investigated whether MEKi might have affected the development of memory phenotypes in CD8+ T cells. Within MEKi-treated T cells, they observed markers of memory generation and identified populations of central memory-like (CD62L+CD44+) and naive-like (CD62L+CD44-) CD8+ T cells; however, within the naive-like population, many cells expressed high levels of memory markers, including CD95 and CCR7, in line with recent observations of a population of minimally differentiated stem cell-like memory T (TSCM) cells. Expression of IL-2Rβ provided additional evidence of stem-like characteristics in these cells, suggesting that MEKi enriches CD8+ TSCM cells in tumors.

Next, the researchers characterized these cells in vitro by activating CD8+ T cells with gp100. Without MEKi, most of the CD8+ T cells attained a TCM phenotype, while a minor portion remained naive. When MEKi was added, a large portion of CD8+ T cells attained a TSCM phenotype; these cells were naive-like and showed lower mitochondrial potential, higher proliferative capacity, higher expression of the stem cell/T cell memory marker Sca1, higher expression of activation and memory markers, lower expression of effector and exhaustion markers, and higher expression of KLF2, which is associated with self-renewability, prolonged survival, and reduced apoptosis. Further studies showed that simultaneous inhibition of both MEK1 and MEK2 was required for TSCM generation.

To further establish that TSCM cells were distinct from Tnaive and TCM cells, Verma et al. analyzed human MEKi-treated CD3/CD28-activated CD8+ T cells. Based on methylation profiles and CpG DNA methylation of key genes dynamically regulated during T cell memory development, the researchers showed that MEKi-induced TSCM cells (CD45RA+CCR7+CD95+) were distinct from both TCM cells (CD45RA-CCR7+) and Tnaive cells (CD45RA+CCR7-CD95-), and were most similar to freshly isolated bona fide TSCM cells. The phenotypes were also functionally differentiated by their multipotency/plasticity proliferation indices and self-renewal capacities. Based on their observations, Verma et al. determined that the TSCM phenotype is an intermediate between Tnaive and TCM phenotypes.

To determine when and how MEKi induces the generation TSCM cells, Verma et al. pre-treated naive T cells with gp100, then later treated them with MEKi. This failed to induce the TSCM cell generation seen when gp100 and MEKi were administered simultaneously, suggesting that MEKi acts during TCR-mediated cell priming. Looking at cell cycle progression and differentiation, the researchers found that MEKi delayed cell cycle progression, leading to an accumulation of activated cells in earlier stages of cell division. MEKi did not interfere with PI3K- or Akt-mediated T cell activation, suggesting that it inhibits the cell cycle at an early phase while allowing for antigen-mediated activation. By knocking out or inhibiting various molecules along different pathways, Verma et al. showed that both the delay in cycle progression and the enhanced metabolic fitness induced by MEKi contributed to the generation of TSCM cells, and that both effects were required for optimal TSCM cell generation.

Testing whether MEKi treatment might be able to enhance CD8+ T cells for adoptive cell transfer (ACT), Verma et al. found that transfer of MEKi-treated, gp100-stimulated pMEL-1 CD8+ T cells prepared in vitro showed stronger effector recall responses than controls stimulated with gp100 alone, producing higher levels of IFNγ and granzyme B after antigenic rechallenge. ACT of gp100- and MEKi-treated cells into mice bearing B16F10 melanoma tumors resulted in a stronger antitumor response and prolonged survival compared to ACT of CD8+ T cells activated with gp100 alone. To determine the contribution of TSCM cells, the researchers depleted the cell product of CD62L+CD44-Sca1+ cells and repeated the experiment. While the gp100-treated cells showed similar efficacy upon adoptive cell transfer, the gp100- and MEKi-treated, TSCM-depleted cell product was less effective. Interestingly, these cells were still more effective than those treated with gp100 alone, suggesting that MEKi also enhances the antitumor efficacy of TCM cells. Long term, adoptively transferred MEKi-treated cells maintained a memory phenotype more than non-treated cells.

Overall, Verma et al. showed that MEKi enhances the generation of TSCM cells by enhancing metabolic fitness and delaying cell cycle progression. MEKi could be particularly useful in enhancing CD8+ T cell products for ACT, as MEKi-treated T cells were more effective against tumors and prolonged survival in mice.

by Lauren Hitchings

Meet the researcher

This week, lead author Samir Khleif answered our questions.

What prompted you to tackle this research question?

T cell differentiation is highly important for proper immunologic responses. Accordingly, understanding the mechanism that regulates such differentiation is crucial not only to properly appreciate the fundamental biology of the T cells, but also to be able to develop effective strategies for immunotherapy of cancer. Furthermore, stem cell memory CD8+ T cells (TSCM) are highly potent and regenerative T cells. Understanding the signaling pathways that control the generation of TSCM cells would provide effective tools to pharmacologically reprogram CD8+ T cells at the appropriate time and conditions into this highly effective “super” T cell, and hence generate more effective immune therapeutic approaches.

What was the most surprising finding of this study for you?

The ability of the MEK pathway to highly and effectively control the differentiation of CD8+ T cells towards terminal differentiation, especially by enhancing their metabolic fitness. Also surprising was the high potential for such reprogramming to enhance the immunotherapy of cancer in a direct pharmacologic, therapeutic approach, and/or to enhance the potency of CD8+ ex vivo for use in an adoptive T cell approach.

What was the coolest thing you’ve learned (about) recently outside of work?

We may be able to do many things from a distance as effectively, or in some cases more effectively than in-person; however, human contact – discussion over a dinner or a drink – especially in science is highly missed as part of the creative cycle of our lives. This last year has been very special by all measures; however, one of the coolest things that we have lived through as scientists is the amazing dedication of our community, and the remarkable international collaboration leading to historical unprecedented discoveries. That was soooo COOL!

References:

Verma V., Jafarzadeh N., Boi S., Kundu S., Jiang Z., Fan Y., Lopez J., Nandre R., Zeng P., Alolaqi F., Ahmad S., Gaur P., Barry S.T., Valge-Archer V.E., Smith P.D., Banchereau J., Mkrtichyan M., Youngblood B., Rodriguez P.C., Gupta S., Khleif S.N. MEK inhibition reprograms CD8+ T lymphocytes into memory stem cells with potent antitumor effects. Nat Immunol. 2020 Nov 23. 

In the Spotlight...

Shared Immunogenic Poly-Epitope Frameshift Mutations in Microsatellite Unstable Tumors

By analyzing tumor whole exome sequencing data in TCGA and querying the Cancer Cell Line Encyclopedia, Roudko and Bozkus et al. showed that microsatellite instability-high (MSI-H) endometrial, colorectal, and stomach cancers had high recurring frameshift mutation loads that frequently encoded polyepitope neopeptides that bound to MHC alleles prevalent in patients with MSI-H cancers, more so in anti-PD-1 responders. Analysis of The Immune Epitope Database showed these tumor neoantigens were distinct from self and viral antigens. Derived, predicted neopeptides stimulated blood CD8+ T cells from healthy donors and patients to make IFNγ and TNFα.

Contributed by Paula Hochman

Eftilagimod alpha, a soluble lymphocyte activation gene-3 (LAG-3) protein plus pembrolizumab in patients with metastatic melanoma

Atkinson et al. report the safety and efficacy of agonistic soluble LAG3 protein (Eftilagimod alpha; efti) in combination with anti-PD-1 (pembrolizumab) in patients with unresectable or metastatic melanoma. In vitro, efti was able to enhance antigen-induced human T cell responses, and in vivo, efti was well tolerated in combination with pembrolizumab without any dose-limiting toxicity. All patients treated with 30 mg efti maintained effective plasma concentration and showed a sustained increase in activated CD4+ and CD8+ T cells. The immune-related (ir) overall response rate was 54% and ir-disease control rate (irDCR) was 75%.

Contributed by Shishir Pant

An engineered 4-1BBL fusion protein with "activity on demand"

Mock et al. generated a panel of cytokine–antibody fusion proteins comprising different formats of murine 4-1BB ligand and the antibody F8, which binds to the human (and mouse) tumor-associated, alternatively spliced EDA domain of fibronectin. The top candidate, F8–4-1BBL was inactive in solution, but regained agonist 4-1BB activity upon binding EDA. In vivo biodistribution studies showed that F8-4-1BBL selectively localized to EDA+ tumor vasculature with no binding to normal tissues. In tumor models, the fusion protein exhibited potent antitumor efficacy alone and in combination with PD-1 blockade, without apparent toxicity.

Contributed by Katherine Turner

Combining T-cell-specific activation and in vivo gene delivery through CD3-targeted lentiviral vectors

To overcome the complexity and cost of production of autologous CAR constructs, Frank et al. demonstrated that lentiviruses containing scFvs that simultaneously target and activate CD3 on T cells (LV-CD3) efficiently and specifically activate and transduce human CD4+ and CD8+ T cells in vitro, in whole blood, and in vivo (humanized mice). Off-target (CD45- or CD45+CD19+) transduction was minimal. In humanized mice, LV-CD3-CD19  constructs efficiently generated CD19-CARs that were durable and that depleted CD19+ B cells; both results were enhanced if mice were pretreated with IL-7, which preferentially stimulates naive and memory cells.

Contributed by Ed Fritsch

Concurrent Dexamethasone Limits the Clinical Benefit of Immune Checkpoint Blockade in Glioblastoma

Iorgulescu et al. evaluated the impact of dexamethasone on response to anti-PD-1 therapy in glioblastoma (GBM) in immune-sensitive GL261 and immune-resistant CT-2A GBM intracranial murine models. Concurrent dexamethasone administration decreased the survival benefit of anti-PD-1 therapy, with or without radiotherapy, in a dose-dependent manner. Dexamethasone increased apoptosis, decreased activation of  CD4+ and CD8+ T cells, and decreased intratumoral and systemic CD4+ T cell, CD8+ T cell, and NK cell populations in vivo. In patients with IDH wild-type GBM undergoing anti–PD-(L)1 therapy, dexamethasone use was associated with poor survival.

Contributed by Shishir Pant

Antitumor activity without on-target off-tumor toxicity of GD2-chimeric antigen receptor T cells in patients with neuroblastoma

In a phase I clinical trial, 12 young neuroblastoma patients were treated with escalating doses of GD2-targeted, kill switch-containing second generation CAR T cells and lymphodepletion. CAR T cells expansion was observed only in patients with high CAR doses (≥108 cels/m2) and prior lymphodepletion, with no dose-limiting toxicities. Although objective clinical responses did not occur, three patients in the high-dose cohort showed evidence of persistent immune activation and antitumor CAR T cell activity, particularly in bone marrow and soft tissue lesions. In post-treatment biopsies, GD2 antigen expression was maintained, but CAR T cells were not detected.

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.