Weekly Digests
‹ Back to October

A new internal T cell checkpoint: CISH

October 14, 2020

PD-1 and other known markers of activation and exhaustion have helped researchers to understand T cell biology and develop numerous immunotherapies. While many of these markers are well known and well studied, there are likely more that have yet to be investigated. Looking into the relationship between PD-1 and other markers of activation, Palmer and Webber et al. noticed something interesting about the expression patterns of CISH (cytokine-induced SH2 protein) and did a deep dive into understanding the biology of what could be an important internal checkpoint regulating T cell reactivity. A pre-print version of their findings, which have not yet been peer reviewed, was recently published on bioRxIV. 

While studying markers of activation and exhaustion on fresh TIL from treatment-naive melanoma patients, Palmer and Webber et al. noticed that cells expressing activation markers such as PD-1, 4-1BB, TIM3, TOX, and CD39 were distinct from cells expressing CISH. In fact, PD-1 and CISH appeared to be almost mutually exclusive. Further, while CISH+CD8+ T cells were fairly uncommon in peripheral blood samples, they were enriched in tumors. Looking at naive T cells, central memory T cells, and effector memory T cells, the researchers found that each expressed low levels of CISH before TCR stimulation and upregulated CISH significantly following TCR stimulation. This effect was more pronounced with advancing differentiation status, suggesting that CISH expression is induced by TCR stimulation and increases with chronic antigen stimulation. 

To investigate the functional significance of CISH expression, the researchers developed a CRISPR/Cas9-based strategy to knock out expression of CISH in human T cells. Upon anti-CD3-mediated TCR stimulation, CISH knockout (KO) cells showed increased production of IFNγ, TNFα, and IL-2, and a higher portion of CISH KO cells were polyfunctional compared to wild-type (WT) cells. These results suggest that CISH KO enhanced functional activation after TCR stimulation.

Next, Palmer and Webber et al. engineered CISH KO T cells with an HLA-A2-restricted TCR specific for NY-ESO-1 to evaluate the role of CISH in antigen responses. In coculture with NY-ESO-1+ cells, a higher portion of CISH KO compared to WT cells were polyfunctional – an increase not observed in coculture with NY-ESO-1- cells. This pattern was maintained when CISH KO cells were deployed in an assay against relevant and irrelevant tumors, suggesting that CISH inhibits TCR stimulation in an antigen-specific manner.

Based on this evidence, the researchers hypothesized that CISH KO may be able to restore weak or suppressed T cell responses to neoantigens by enhancing their reactivity to neoantigen-driven TCR stimulation. To study this, they developed a reagent-sparing strategy that allowed them to knock out CISH in TILs prior to ex vivo rapid expansion (REP). The team utilized samples from a patient with metastatic colon cancer that expressed an identifiable neoantigen and contained T cells that were reactive to that neoantigen. When CISH KO TILs were cultured with autologous DCs loaded with the neoantigen, more cells produced IFNγ, TNFα, and/or IL-2 than their WT counterparts. This was maintained even when low levels of the neoantigen were loaded, suggesting that CISH KO enhances both T cell functionality and sensitivity to antigenic stimulation. Similar results were observed using samples from a second patient, suggesting reproducibility. 

Having shown that CISH KO could enhance the sensitivity of a T cell response to a neoantigen, the researchers wanted to know whether CISH KO could resurrect the functionality of neoantigen-specific T cells that had lost their ability to respond following REP. Using samples from a patient with TILs that were reactive to a neoantigen, but lost their reactivity after a REP, they found that while control TILs showed no reactivity to the neoantigen following REP, CISH KO TILs responded to neoantigen peptide-loaded DCs. This suggested that the loss of reactivity after traditional TIL REP could be restored with CISH KO.

Next, Palmer and Webber et al. wondered whether CISH might play a role in dampening immune responses to common neoantigens, like those that arise from TP53 driver mutations. Using 18 tumor samples with TP53 mutations and mutant p53 neoantigens, 7 of which showed evidence of T cell reactivity, the researchers found that genes associated with T cell activation were expressed in the reactive samples, while CISH expression clustered with non-reactive samples. When CISH was knocked out in a sample of TILs from a patient with a modest response to mutant p53, CISH KO TILs dramatically increased expression of IFNγ in response to mutant p53-loaded DCs, and more of the cells were polyfunctional compared to the unaltered TILs.

Interestingly, while CISH KO cells showed evidence of hyperactivation, they did not show signs of accelerated maturation, which would typically be expected. Investigating this unusual finding, the researchers found that PLC-γ1 and SLP76, intermediate molecules involved in TCR signaling, were phosphorylated with increased intensity and duration after TCR stimulation in CISH KO cells. Subsequently, there was an increase in the amplitude and duration of NFAT and NFκB translocation to the nucleus, a slight increase in c-Jun, and enhanced phosphorylation of ERK. However, enhanced phosphorylation of AKT, which is largely associated with T cell maturation, was not observed. 

Given that CISH expression and PD-1 expression were initially found to be mutually exclusive, Palmer and Webber et al. evaluated whether CISH plays a role in the regulation of exhaustion markers and found that low CISH expression was associated with increased PD-1. In mice with gp-100-expressing melanoma tumors treated with adoptively transferred WT or CISH KO gp-100-specific pmel-1 cells, CISH KO cells were found to express higher levels of PD-1 eight days after adoptive transfer, suggesting potential synergy with PD-1 blockade. Comparing treatment combinations, the researchers found that while CISH KO pmel-1 cells alone or WT pmel-1 cells plus anti-PD-1 slowed tumor growth, CISH KO pmel-1 cells plus anti-PD-1 resulted in profound tumor regression and long-term survival of mice.

Together these results showed that CISH acts as an internal regulator of T cell reactivity, dampening responsiveness to neoantigens. CISH KO increased T cell sensitivity to neoantigens and enhanced their activation, functional responses, and tumor-killing capacity. CISH KO could potentially be used to enhance TIL therapy or improve responses to PD-1 checkpoint blockade, and warrants further study as a target for immunotherapy.

by Lauren Hitchings

Meet the researcher

This week, we asked first co-author Douglas Palmer to answer the following questions.

First co-author Douglas Palmer with co-author (and wife) Zulmarie Franco, in Patagonia.

What prompted you to tackle this research question?
Understanding the role of negative regulators in T cell immunity has long been a passion of mine [1]. It began when we noticed that T cells acutely stop killing tumors after adoptive cell transfer [2] and I wanted to understand why and how we could reverse it. Unfortunately, at the time, genetic manipulation of these negative regulators in fully mature T cells was in its infancy and I was relegated to evaluating their role in tumor immunity using mouse knockout models and siRNA. What I uncovered though, was that the disruption of the Suppressor of Cytokine Signaling (SOCS) member, CISH, could result in profound and long-lasting tumor-free survival in animal models [3]. Surprisingly, I uncovered that this molecule blocked T cell receptor signaling and not cytokine signaling, as was previously thought at the time.
It is known that tumor-resident T cells (TIL) have neoantigen specificity for the tumors they reside in, however the tumor still grows, and the adoptive transfer of these cells fails to reliably result in tumor regression. I wanted to know how CISH related to other known markers of activation/exhaustion and if the deletion in fully mature TILs would restore their tumor killing capabilities. The advent of CRISPR and chemically-modified guide RNAs [4] was the big game changer that made this study possible.

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

What was most surprising was that in TILs, I found that CISH was inversely expressed with known activation/exhaustion markers like TOX, CD39, and PD-1. It appeared that CISH might inhibit their expression and the knockout of CISH resulted in upregulation of PD-1, which I exploited using a combinatorial approach.

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

I’m an avid sailor and love to race. A few years back I took part in an ocean race from Annapolis, MD to St. George, Bermuda. There were about 40 other boats and I was crewing on a 50 ft sloop with 6 other guys. Our crew of seven was a motley bunch, all different demeanors and skill sets. One guy, an ex-Navy fighter pilot, was an excellent navigator. Another was a structural engineer and excellent at repairing broken sails. My specialty was sail aerodynamics, in particular spinnakers, which I trimmed for days on end. After we left the shelter of the Chesapeake Bay and entered the ocean, it was just the seven of us and we never saw another boat up until we got the reefs on the north shore of Bermuda six days later. Teamwork was key. Teamwork, getting along, and playing to our strengths got us through several big storms, ocean swells and eddies, and broken equipment. Teamwork got us safely to Bermuda and ultimately second place on the ocean portion of the race. I think understanding that concept will not only help get you across an ocean in a floating tub of plastic, but is central in developing new therapies to improve the lives of cancer patients.

1. Palmer, D.C. and N.P. Restifo, Suppressors of cytokine signaling (SOCS) in T cell differentiation, maturation, and function. Trends Immunol, 2009. 30(12): p. 592-602.
2. Palmer, D.C., et al., Effective tumor treatment targeting a melanoma/melanocyte-associated antigen triggers severe ocular autoimmunity. Proc Natl Acad Sci U S A, 2008. 105(23): p. 8061-6.
3. Palmer, D.C., et al., Cish actively silences TCR signaling in CD8+ T cells to maintain tumor tolerance. J Exp Med, 2015. 212(12): p. 2095-113.
4. Hendel, A., et al., Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells. Nature Biotechnology, 2015. 33(9): p. 985-989.

References:

Douglas C. Palmer, Beau R. Webber, Yogin Patel, Matthew J. Johnson, Christine M. Kariya, Walker S. Lahr, Maria R. Parkhurst, Jared J Gartner, Todd D Prickett, Frank J. Lowery, Rigel J. Kishton, Devikala Gurusamy, Zulmarie Franco, Suman K. Vodnala, Miechaleen D. Diers, Natalie K. Wolf, Nicholas J. Slipek, David H. McKenna, Darin Sumstad, Lydia Viney, Tom Henley, Tilmann Bürckstümmer, Oliver Baker, Ying Hu, Chunhua Yan, Daoud Meerzaman, Kartik Padhan, Winnie Lo, Parisa Malekzadeh, Li Jia, Drew C. Deniger, Shashank J. Patel, Paul F. Robbins, R. Scott McIvor, Modassir Choudhry, Steven A. Rosenberg, Branden S. Moriarity, Nicholas P. Restifo. Internal checkpoint regulates T cell neoantigen reactivity and susceptibility to PD1 blockade. bioRxiv 2020.09.24.306571; 
This article is a pre-print and has not been certified by peer review
.

In the Spotlight...

Exosomes derived from Vδ2-T cells control Epstein-Barr virus–associated tumors and induce T cell antitumor immunity

Wang et al. demonstrated that Vδ2-T cell-derived exosomes (Vδ2-T-Exos) induced EBV tumor cell apoptosis and activated CD4+ and CD8+ T cell–mediated antitumor immunity. Vδ2-T-Exos contained death-inducing ligands FasL and TRAIL, NKG2D, MHC class I and II, CD80, and CD86. Vδ2-T-Exos required NKG2D to enter EBV tumor cells, and FasL and TRAIL ligands to induce apoptosis. Vδ2-T-Exos controlled EBV-associated tumors in Rag2−/−γc−/− and humanized mice. Allogeneic Vδ2-T-Exos enhanced T cell recruitment, activation, and expansion, and demonstrated superior EBV tumor control over autologous Vδ2-T-Exos, DC-Exos, and NK-Exos.

Contributed by Shishir Pant

Regulatory T cell control of systemic immunity and immunotherapy response in liver metastasis

Lee et al. investigated why cancer patients with liver mets have worse outcomes following anti-PD-1 therapy compared with those without liver mets. A dual-tumor (s.c. and liver) mouse model showed that a liver tumor antigen generated an immune response that led to systemic antigen-specific suppression in the s.c. tumor (not observed in a s.c. and lung model). Suppression was due to liver-specific Treg activation resulting in modulation of i.t. antigen-expressing CD11b+ monocytes and subsequent suppression of CD4+ and CD8+ effector T cells in the s.q. site. Suppression could not be reversed by anti-PD-1 unless Tregs were depleted or destabilized.

Contributed by Katherine Turner

Blockade of immune checkpoints in lymph nodes through locoregional delivery augments cancer immunotherapy

In mice bearing B16F10 tumors, Francis et al. observed that i.t. rather than i.p. administration of anti-PD-1 and anti-CTLA-4 (ICB) mAbs extended survival, increased CD8+ T cell tumor infiltration, and boosted CD8+ T cell proliferation both in tumors and tumor-draining lymph nodes (TDLNs). Compared to i.p., local (i.t. or ipsilateral intradermal) injection improved TDLN mAb accumulation and efficacy in multiple tumor models. Local delivery was effective at lower doses, was independent of mAb capability to deplete Tregs, and reduced toxicity biomarkers. A hydrogel formulation sustained ICB release, increasing LN delivery and tumor control.

Contributed by Alex Najibi

An implantable blood clot-based immune niche for enhanced cancer vaccination

Fan and Ma et al. developed a vacuum-treated autologous blood clot vaccine (BCV) consisting of tumor antigens, TLR9 agonist (CpG-ODN), and GM-CSF to induce anticancer immune responses. BCV recruited and activated proinflammatory immune cells to create an immune niche with increased cytokine and stimulatory molecule expression in vivo. In combination with anti-PD-1, BCV demonstrated prophylactic efficacy against B16-OVA, as well as antitumor immunity against established B16F10 and 4T1 tumors. This combination also inhibited postsurgical recurrence, with increases in IFN-γ+ and Ki67+ cytotoxic CD8+ T cells and NK cells.

Contributed by Shishir Pant

An antibody targeting ICOS increases intratumoral cytotoxic to regulatory T cell ratio and induces tumor regression

Using TCGA datasets, scRNAseq, and immunostaining, Sainson and Thotakura et al. showed that ICOS expression in tumor-bearing mice and patients with cancers was highest on intratumoral Tregs. An ICOS-specific human monoclonal IgG1 made in Icos-/- transgenic mice bound human, monkey, rat, and mouse ICOS, killed ICOShi T cells via ADCC, and induced IFNγ and TNFα secretion by ICOSlo TEFF cells in human cells in vitro. Anti-ICOS IgG1 treatment of monkeys or mice (reformatted as a murine IgG2a) depleted ICOShi T cells, and in syngeneic mouse models, blocked lymphoma growth, improved anti-PD-L1 efficacy in models of solid tumor, and increased the intratumoral TEFF:Treg ratio.

Contributed by Paula Hochman

Reactive Myelopoiesis Triggered by Lymphodepleting Chemotherapy Limits the Efficacy of Adoptive T Cell Therapy

Innamarato et al. show that after cyclophosphamide-fludarabine (Cy/Flu) lymphodepleting chemotherapy, immunosuppressive CD11b+ myeloid cells accumulated in peripheral blood, which correlated with a decrease in tumor-specific T cell persistence and poor PFS and OS in melanoma patients. Cy/Flu lymphodepletion mobilized myeloid progenitor cells, expanded MDSCs, and increased IL-6 expression in bone marrow-derived cells in the B16 melanoma model. IL-6 blockade reduced the suppressive capacity of MDSCs, increased Fas expression, sensitized MDSCs to apoptotic signals, and enhanced ACT efficacy in vivo.

Contributed by Shishir Pant

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.