Weekly Digests
‹ Back to May

CTLA-4 and PD-1 set the limits on T cell differentiation

May 15, 2019

Positive and negative costimulatory molecules on T cells are known to play a role in T cell activation, however, whether they play a role in T cell differentiation (along with TCR signal strength and cytokine signaling) is not as well defined. Following observations that genetic loss or blockade of CTLA-4 can affect CD4+ T cell phenotypes, Wei and Sharma et al. investigated the role of negative regulators CTLA-4 and PD-1 in T cell differentiation and determination of phenotype; their findings were recently published in Immunity.

To better understand the role of CTLA-4 in T cell differentiation, Wei and Sharma et al. profiled lymphocytes from a Ctla4-/- mouse model via mass cytometry, and conducted unsupervised clustering to identify phenotypes. Compared to wild type Ctla4+/+ and heterozygous Ctla4+/- controls, the distribution of phenotypes across lymphocytes from Ctla4-/- was distinct. Within both the CD8+ and CD4+ T cell compartments, multiple phenotypes appeared to be enriched in Ctla4-/- mice, including terminally differentiated CD8+ T cells and an ICOS+ Treg population that was very minor in control mice. More interestingly though, some phenotypes within the CD4+ compartment were entirely unique to the Ctla4-/- mice, including populations of unskewed ICOS+ cells, ICOS+ Th1-skewed Tregs, ICOS+ Th1-like effectors, and Th17-like cells. The emergence of these unique, non-canonical CD4+ T cell subsets in the absence of CTLA-4 expression suggests that CTLA-4 signaling limits the boundaries of phenotypic expression within the CD4+, but not the CD8+ T cell compartment, in peripheral lymph node tissues. Archetypal analysis (a computational approach) confirmed these findings and defined these distinct new cell types as new archetypes with a significantly different set of phenotypes not observed in control mice.

In an effort to understand how these non-canonical phenotypes arise in the absence of CTLA-4, Wei and Sharma et al. investigated a number of factors. First, the researchers investigated whether unrestricted activation and proliferation may have contributed to the novel phenotype populations. To this end, CTLA4-/- and control were vaccinated with ovalbumin to induce a strong T cell response against a number of antigens, and while some T cell phenotypes did expand due to the enhanced T cell activation, the novel phenotypes did not, indicating that their emergence was not an indirect effect of CTLA-4 loss by enhanced T cell activation. Next, the researchers examined the high ICOS expression that was shared across the novel phenotypic subsets and found that while ICOS expression was essential to the functionality of these cells, it did not play a role in driving their development. Finally, the researchers analyzed the TCRβ repertoire in thymic and lymph node tissue to determine whether changes in the central or peripheral repertoire might underlie differentiation towards novel CD4+ T cell phenotypes. While no major differences in the thymic TCR repertoire were observed, T cell clonality was increased in lymph node tissue in Ctla4-/- mice. Interestingly, specific self-reactive T cell clones were found to be recurrently expanded or in a few cases, contracted, across Ctla4-/- animals, and this at least partially influenced differentiation towards atypical CD4+ T cell phenotypes.

In order to identify specific molecular events that are restricted by the expression of CTLA-4 in T cells, the researchers computationally constructed T cell differentiation pathways based on quantitative mass spectrometry data. Changes in key marker expression could be observed tracking differentiation from a naive-like state and resulting in the discrete archetypes observed only in CTLA-4-deficient mice. The researchers found that while CTLA-4 does not keep any pathways fully dormant, it does impose maximum limits on the expression levels of active pathways. Genetic absence of CTLA-4 therefore allowed cells to extend beyond the phenotypic boundaries of their destined cell fate. Ratios of key transcription factors (e.g. FoxP3, RORγT, TBET, GATA3, BCL6) changed when a normal differentiation boundary was approached. Analyzing cytokine production by T cells from Ctla4-/- mice in comparison to controls, the researchers identified an increased frequency of cytokine-producing cells and increased expression of IFNγ, TNFα, and IL-17α, particularly by ICOS+ cells. This indicated that loss of CTLA-4 increased the frequency of functionally active CD4+ effector cells. Genetic loss of Ctla4 also led to an increased frequency of cytokine-producing Tregs, however, it did not enhance their cytokine production capacity, and in fact decreased their suppressive functionality.

To determine whether the capacity to restrict phenotypic boundaries within T cells applied to negative costimulatory molecules beyond CTLA-4, the researchers turned to PD-1. In contrast to observations in Ctla4-/- mice, the phenotypic boundaries of CD4+ T cells were not expanded in Pdcd1-/- mice compared to controls, whereas the phenotypic boundaries of CD8+ T cells were somewhat expanded (though this effect was only seen in older mice). These results were consistent with a model in which CTLA-4 regulation is induced immediately after T cell priming, while PD-1-mediated regulation is engaged only after the initiation of an active immune response. Based on this, it is likely that both CTLA-4 and PD-1 impose phenotypic boundaries in T cells, but they do so through different mechanisms and in different T cell subsets, and act to a different extent.

Next, the researchers naturally wondered whether antibody blockade of CTLA-4 or PD-1 had similar effects to genetic deletion of these negative costimulators. They found that CTLA-4 blockade enriched T cells for a T-BET+TCF1+CD4+ T cell archetype, and that it did not enrich any CD8+ T cell archetype. Meanwhile, PD-1 blockade enriched a PD-1+PD-L1intCD8+ T cell archetype. Unlike in the genetic studies, however, none of the enriched archetypes were unique compared to controls. This less dramatic phenotypic expansion is likely attributed to incomplete loss of signaling from antibody blockade, compared to the complete loss of signaling afforded by genetic engineering.

Overall, the evidence uncovered by Wei and Sharma et al. shows that CTLA-4 and PD-1, in addition to their effects on activation, impose boundaries on T cell phenotypes during peripheral differentiation. T cells engineered to lack these molecules are therefore able to differentiate further down active pathways, leading to the acquisition of phenotypes not observed under natural circumstances. Similarly, antibody blockade of these molecules may similarly reduce some of the limitations on T cell differentiation.

by Lauren Hitchings

Meet the Researcher

This week, Spencer C. Wei and Roshan Sharma, first co-authors on this study, answered our questions.

(Spencer C. Wei, first co-author)

What prompted you to tackle this research question?
There is so much that we still do not know about how T cell activity is regulated and insights into basic biological processes are absolutely essential for improving immunotherapy approaches. Here we wanted to understand how T cell phenotypes are defined and whether T cell costimulation plays a role in this process.

What was the most surprising finding of this study for you?
How profound of a role CTLA-4 plays in regulating CD4+ T cell phenotypic limits. This function complements, but is distinct from its well-known role in regulating T cell activation. This changes our perspective of how we think about how CTLA-4 regulates and shapes T cell biology.

Roshan Sharma (first co-author) and Dana Pe'er (senior co-author)

What prompted you to tackle this research question?
The primary question we tackle in this project can be stated as "Do negative costimulatory molecules play a role in T cell differentiation?". We arrived at this question with inspiration and motivation from an earlier collaboration between the Pe'er and Allison lab, which utilized mass cytometry data and machine learning tools to dissect the cellular mechanisms of anti-CTLA-4 and anti-PD-1 treated tumor-infiltrating T cells. The study revealed that anti-CTLA-4 and anti-PD-1 checkpoint blockade act through distinct cellular mechanisms. In particular, it was found that anti-PD-1 treatment resulted in an expansion of exhausted CD8+ T cells subsets while anti-CTLA-4 treatment resulted in a predominant expansion of Th1-like CD4+ T cell subsets. In addition, highlighting the cellular underpinnings of checkpoint blockade, the study hinted at the fact that molecules such as CTLA-4 and PD-1, also known as negative costimulatory molecules, might play a role in T cell differentiation along with their well-known and essential role in T cell activation. Therefore, we wanted to take a more basic approach to target this question directly.

What was the most surprising finding of this study for you?
The most interesting conclusion of the study was that negative costimulatory molecules indeed play a role in T cell differentiation in addition to their role in T cell activation. What was surprising was the way they seem to influence differentiation, which is by putting restriction on the differentiation limits of the T cells. Essentially, our work highlights that CTLA-4 and PD-1 act as "gate-keepers", which restrict the expansion of phenotypic states of differentiating T cells. Once they are deleted, the cells can expand along a continuum to occupy phenotypic states not attainable under normal circumstances. Further analysis led us to observe that protein expression behavior is largely maintained intact despite the loss of CTLA-4, that is, our study argued against the possibility of activation of otherwise dormant signaling pathways under normal circumstances. In summary, loss of negative costimulatory molecules allow normal signaling behavior but at an exaggerated or expanded level.

What was the coolest thing you’ve learned (about) recently outside of the lab?
I recently learned about a paper titled "A network approach to topic models" from a colleague. The paper essentially presents a marriage of network science with topic models, typically thought of as a distinct branch of computer science.

References:

Wei S.C., Sharma R., Anang N.A.S., Levine J.H., Zhao Y., Mancuso J.J., Setty M., Sharma P., Wang J., Pe'er D., Allison J.P. Negative Co-stimulation Constrains T Cell Differentiation by Imposing Boundaries on Possible Cell States. Immunity. 2019 Apr 16.

In the Spotlight...

REVIEW: The promise of neoadjuvant immunotherapy and surgery for cancer treatment

O’Donnell and Hoefsmit et al. argue that neoadjuvant immunotherapy may be superior to adjuvant immunotherapy in combination with surgery. Preclinical and limited clinical evidence supports the hypothesis that inducing an antitumor immune response while the primary tumor mass is present increases the magnitude, breadth, and durability of response, preventing metastatic relapse, though more directly comparable data is needed. The review also addresses the need to optimize treatment timing (to prevent T cell re-exhaustion or surgical delay due to adverse events), agents, and monitoring strategies to maximize long-term clinical response.

Costimulation Induces CD4 T Cell Antitumor Immunity via an Innate-like Mechanism

Morales Del Valle et al. developed a mouse model chronically exposing a tumor-unrelated antigen on MHC-II+CD11c+ DCs, and showed that antigen-specific CD4+ T cells can differentiate into effectors in the presence of dual costimulation with OX40 and 4-1BB agonists. These antigen-specific CD4+ T cells retained their therapeutic helper function, expressed IFNγ in a TCR-independent manner upon stimulation with a JAK/STAT activating cytokine (IL-2 or IL-12) plus an IL-1 family cytokine (IL-33 or IL-36), and, upon adoptive transfer, conferred an antitumor response in the presence of intratumoral IL-36 and subsequent dual costimulation.

PD-1+ regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer

After 4 of 36 gastric cancer patients treated with anti-PD-1 developed hyperprogressive disease (HPD), Kamada, Togashi, and Tay et al. analyzed the pre-treatment and post-treatment tumor samples and discovered that the treatment increased the frequency of tumor-infiltrating, proliferating, activated PD-1+CD45RA-CD25highFoxP3high effector Tregs in HPD patients. The blockade or deficiency of PD-1 in Tregs increased their proliferation and immunosuppressive function in vitro and in vivo, and promoted tumor growth in mice with B16F0 melanoma. Thus, agents that deplete PD-1+ Tregs during anti-PD-1 treatment may prevent HPD.

Differential effects of depleting versus programming tumor-associated macrophages on engineered T cells in pancreatic ductal adenocarcinoma

Stromnes et al. combined mesothelin-specific CD8+ T cell (TCRMSLN) transfer with two tumor-associated macrophage (TAM)-targeting strategies in murine pancreatic ductal adenocarcinoma (PDA). TCRMSLN cells alone increased intratumoral M1 TAMs and survival. Anti-CSF1R decreased M2 TAMs and improved intratumoral endogenous CD8+ T cell numbers but had minimal impact on TCRMSLN cells. Agonist-CD40 boosted TCRMSLN cell persistence and Ki67/GzmB levels, reduced PD-1 expression, and supported remodeling of the tumor stroma, but did not rescue IFNγ production. In human PDA, M2 TAMs correlated with CSF1/CSF1R expression.

Contributed by Alex Najibi

CLINICAL TRIAL: Rational design of anti-GITR-based combination immunotherapy

In a phase I clinical trial, 43 patients with advanced cancers were treated with a single dose of TRX518, an agonistic monoclonal antibody that triggers GITR signaling, inducing hyperactivation and destabilization of activated Tregs. Treatment was well-tolerated and reduced peripheral and intratumoral Tregs, however, it did not induce clinical responses. Mouse models suggested that extended exposure to Tregs induces PD-1+ exhaustion in effector T cells, limiting their antitumor efficacy. Combining anti-GITR with anti-PD-1 to reverse cell exhaustion overcame resistance in mice. Early clinical results of the combination are promising.

Engineering the TGFβ receptor to Enhance the Therapeutic Potential of Natural Killer Cells as an Immunotherapy for Neuroblastoma

Burga et al. engineered human umbilical cord-derived NK cells to express a dominant negative TGFβ receptor alone (RBDNR) or coupled with an NK-specific activating domain (either DAP12 [NKA] or synNotch/RELA [NKCT]) to block TGFβ signaling and convert the signal from inhibitory to activating. All NK variants exhibited increased activation and cytolytic activity against human neuroblastoma compared with untransduced NK cells in the presence of TGFβ in vitro; however, only NKA-transduced NK cells conferred superior persistence, protection against tumor progression, and survival in a xenograft human neuroblastoma model in immunodeficient NSG mice.

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.