Weekly Digests
‹ Back to April

Prime time: secondary prolonged DC priming boosts high-affinity T cell responses

April 23, 2025

After initial priming through interaction with dendritic cells (DCs), CD8+ T cells disengage and migrate, but little is known about how cytokine signals are transmitted or received by these cells for differentiation during this migration. Jobin, Seetharama, et al. visualized the dynamics of antigen-specific CD8+ T cells, as well as CD4+ T helper (Th) and regulatory (Treg) cells after initial activation. Their novel findings on a second priming phase were recently published in Science.

The researchers began by assessing in which microenvironments viral antigens are presented by DCs in draining lymph nodes (dLNs) after viral infection. Mice were infected with vaccinia virus (VV) expressing a lymphocytic choriomeningitis virus glycoprotein (VV-GP), and GP-specific TCR transgenic CD8+ T cells (P14) were transferred. Within 24 hours of infection, P14 T cells clustered in the interfollicular area (IFA) and cortical ridge areas of the dLNs, where initial priming took place during long-term interaction with DCs. Three days after infection, P14 cells had proliferated and were distributed throughout the paracortex of the dLNs. Further, cells were actively migrating in the interfollicular area.

To further assess the dynamics, naive P14 cells were transferred at the 3-day time point in this viral model. Eight hours post-transfer, T cells had upregulated CD69 and formed clusters. Antigen-presenting DCs were absent from the IFA, but present in the subfollicular cortical ridge area (T cell zone), where antigen presentation was taking place at day 3 post-infection.

Intravital imaging showed hours-long interactions between the P14 cells and cDC1s in the subfollicular areas. At day 3, large P14 cells (suggestive of lymphoblasts) stopped migrating and interacted with cDC1s for at least an hour. The researchers hypothesized that these secondary tight cellular interactions between T cell blasts and cDC1s in the subfollicular LN area shaped the CD8+ T cell response, and named the process the second priming phase. The T cell blasts interacted with multiple local cDC1s during this phase, while in the initial priming there was a clustering of T cells around one DC. Further, the T cells that re-engaged with DCs proliferated during this period.

To investigate the role of CD4+ T cells during the second priming phase, GP-specific SMARTA CD4+ T cells were co-transferred with P14 CD8+ T cells in a similar experimental setting. SMARTA lymphoblasts were also found in the P14 cell niches, but they showed a different migratory pattern. SMARTA cells stopped migrating and interacted with DCs for approximately 10 minutes, after which they migrated at low speed, moving between DCs. Tregs, on the other hand, did not stop migrating and continued to move at similar speeds as during homeostasis.

Productive immunological synapse formation during antigen-specific cell interactions requires Ca2+ mobilization. In vitro experiments with in vivo-activated P14 cells demonstrated that some cells retained their ability to mobilize Ca2+, indicating that they could respond to TCR stimulation, which is required for cellular arrest. In the mouse model, arrested P14 cells in the subfollicular niches showed Ca2+ flux, suggestive of retention of the capability to respond to TCR signaling at day 3 post-infection.

The researchers then determined which chemokine receptors were involved in the recruitment of T cells to DCs and allowed their long-term interactions. In a model infected with VV-OVA, WT and CXCR3 knockout (KO) OT-1 cells were compared. WT cells arrested migration more commonly and longer than the KO cells, suggesting CXCR3 was important for the migration and retention of CD8+ T cells in the subfollicular hubs during the second priming phase. Further, the KO cells did not differentiate into effector T cells (Teff), while the WT cells did. On day 3, the KO cells had reduced levels of pSTAT5 compared to WT cells, suggesting limited IL-2 signaling.

Since Tregs control immune responses by limiting IL-2 availability, the role of Tregs in this process was further assessed. Treg depletion increased the number of virus-specific OT-1 cells expressing the effector marker KLRG1 on day 8 after VV-OVA infection. Further, OT-1 cells expressed increased levels of pSTAT5 on day 3 postinfection. Further experiments showed that Treg-mediated control of these virus-specific T cell responses did not require CXCR3 expression on the Tregs. Tregs had increased levels of gene expression related to activation of IFN- and IL-2-dependent gene pathways, and the elevated levels of IL-2 signaling signals were primarily observed in “resting” Tregs, a subpopulation found during homeostasis in control lymph nodes with low IL-2 signaling signature. In the absence of Tregs, Teff cell generation from CXCR3 KO OT-1 cells was restored. Therefore, Tregs may limit CD8 effector differentiation during the second priming phase.

The researchers then determined the cellular source of IL-2 during the second priming phase using various models. In mice in which CD8+ T cells were unable to produce IL-2, no difference in effector differentiation was observed after infection. When CD4+ T cells were depleted, OT-1 cells had lower levels of pSTAT5 on day 3 post-infection. IL-2Rα-deficient OT-1 cells had reduced expansion and effector differentiation. In a newly developed model in which only CD4+ Th cells were depleted, but not Tregs, an over 5-fold reduction in the number of virus-specific CD8+ T cells was observed on day 8 after infection. Therefore, CD4+ Th cells may play an important role through production of IL-2 in establishing the quantity of antiviral CD8+ T cell responses.

Finally, the researchers assessed whether the second priming phase provided a mechanism for selecting high-affinity clones for further expansion. Comparing the dynamic behavior of low-affinity OVA-specific CD8+ T cells (OT-3) with their high-affinity counterparts (OT-1) showed limited differences during the initial priming phase in terms of migratory arrest, CD69 expression, and proliferation. However, during the second priming phase, the low-affinity OT-3 cells did not engage long-term with DCs and expressed limited pSTAT5. This resulted in 10-fold fewer OT-3 cells and 100-fold fewer Teff cells on day 8 postinfection.

Together, the data suggest the presence of a second priming phase in which CD8+ T cells re-engage with DCs long-term to get signals for effector differentiation. This process relies on signals from CD4+ Th cells and is regulated by Tregs. These data may provide clues for research into improving the efficacy of vaccines and cellular therapies.

Write-up by Maartje Wouters, image by Lauren Hitchings

Meet the researcher

This week, co-first authors Kasia Jobin and Deeksha Seetharama answered our questions.

From the left: Deeksha Seetharama, Wolfgang Kastenmüller, Kasia Jobin

What was the most surprising finding of this study for you?
We were both very surprised when we noticed that CD8+ T cells re-clustered for a second time after their initial activation. Even more, to see that antigen-specific CD4+ T cells showed a different migration pattern, which again differed from regulatory T cells. Although we had concrete hypotheses about what was happening at this time after the infection, we had to throw them all overboard. It is a great feeling when you observe an unknown process for the first time. Nevertheless, it took us a long time to put all the pieces of the puzzle together, because our results were contradicting paradigms that had long been taken for granted.

What is the outlook?
In principle, we show that the activation of CD8+ T cells is a cyclical process that repeats itself every 2-3 days. After an acute infection, at least in our model, this involves two phases, as the duration of antigen presentation is limited. However, we hypothesize that the principles of cellular interactions which we have discovered for an acute infection also apply to chronic infections and cancer, in which antigen-presentation is continuous. This would have direct implications on treatment regimens for checkpoint immunotherapy or for CAR T cell therapy. We are currently working on precisely these questions. Overall, we are seeing a certain renaissance in research into the role of CD4+ helper T cells in tumor defense, which is only partly based on their cooperation with cytotoxic CD8+ T cells. We believe that the next few years will bring many exciting new findings and therapeutic approaches in this field of research.

What was the coolest thing you’ve learned (about) recently outside of work?
KJ: I’ve been living in Germany for some time and I’m still learning the language. At my home we speak English and Polish. A few nights ago, I was putting my 3-year-old to sleep, and he said: Ohrläppchen. I said: what´s that? He touched my ear lobe and repeated: Ohrläppchen. So, I’ve learned a cool word from a cool little guy.
DS: Recently I travelled to South Korea and learned to properly use chopsticks and make traditional Kimchi. It was exciting to learn their culture and have a well earned break after the paper.

References:

Jobin K, Seetharama D, Rüttger L, Fenton C, Kharybina E, Wirsching A, Huang A, Knöpper K, Kaisho T, Busch DH, Vaeth M, Saliba AE, Graw F, Pulfer A, González SF, Zehn D, Liang Y, Ugur M, Gasteiger G, Kastenmüller W. A distinct priming phase regulates CD8 T cell immunity by orchestrating paracrine IL-2 signals. Science. 2025 Apr 11.

In the Spotlight...

Neoantigen-specific tumor-infiltrating lymphocytes in gastrointestinal cancers: a phase 2 trial

Patients with heavily pretreated metastatic GI cancers were treated with neoantigen-reactive TILs alone (SEL-TIL) or with pembrolizumab (SEL-TIL+P). 15.1% of SEL-TIL and 23.5% of SEL-TIL+P patients had a PR, and 8 additional patients experienced target lesion reduction of >30%. Serious AEs occurred in 30% of patients, with 7 requiring critical care and one infection-related death. Responders’ TIL products were reactive to a significantly greater number of neoantigens and contained a greater number of reactive CD4+, but not CD8+ T cells, while their tumors were enriched for pathways related to inflammation and wound healing.

Contributed by Morgan Janes

Adoptively transferred tumor-specific IL-9-producing cytotoxic CD8+ T cells activate host CD4+ T cells to control tumors with antigen loss

Xiao et al. demonstrated that adoptively transferred Tc9 cells (IL-9-producing cytotoxic CD8+ T cells) were able to control the growth of B16-OVA tumors, even upon antigen loss. Investigating the mechanism, they found that Tc9 cell secretion of IL-24 recruited CCR7+ cDC2s to tumors. These cDC2s migrated to tdLNs, where they primed host cytotoxic effector CD4+ T cells, which then infiltrated tumors and contributed to tumor growth control. In patient data for melanoma and breast cancer, intratumoral IL24 expression correlated with cDC2 and CD4+ T cell signatures, which were both associated with longer survival.

Contributed by Lauren Hitchings

Allogeneic NK cells with a bispecific innate cell engager in refractory relapsed lymphoma: a phase 1 trial

Nieto et al. reported favorable safety profiles and preliminary activity of cord blood-derived cytokine-pre-activated and expanded NK cells pre-complexed with AFM13 – a CD30/CD16A bispecific antibody – in 42 patients with CD30+ lymphoma refractory to both brentuximab vedotin and anti-PD-1. AFM13-NK cell infusion with subsequent infusions of AFM13 were well tolerated, without CRS, neurotoxicity, or GvHD. Donor NK cells persisted up to 3 weeks, acquired an activated and cytotoxic phenotype, and trafficked to tumor sites. At a median follow-up of 20 months, EFS and OS were 26.2% and 76.2%, respectively. Eleven patients remained in CR at 12–40 months.

Contributed by Shishir Pant

Autologous T cell therapy for PRAME+ advanced solid tumors in HLA-A*02+ patients: a phase 1 trial

Wermke et al. reported interim data from a phase 1 dose-escalation trial of IMA203 – PRAME-directed autologous TCR T cell therapy – in HLA-A*02+ patients with PRAME+ recurrent and/or refractory solid tumors. Although the PRAME TCR was pairing-optimized and affinity-enhanced, IMA203 was safe and well tolerated, without any treatment-related fatalities. IMA203 rapidly engrafted tumors and demonstrated long-term persistence. In the 40 patients treated with IMA203, the unconfirmed/confirmed (u/c)ORR was 52.5%, and the cORR was 28.9%, with a median duration of response of 4.4 months. Higher PRAME expression and T cell infiltration correlated with deeper responses and longer PFS.

Contributed by Shishir Pant

Immunotherapy with conventional type-1 dendritic cells induces immune memory and limits tumor relapse

Heras-Murillo et al. treated mice with tumor cell lysate-loaded syngeneic splenic selected cDC populations. cDC1-based vaccines induced cancer-specific Teff and Tmem cells, and reduced tumor burden in therapeutic and prophylactic settings more effectively than cDC2-based vaccines, even in hosts lacking cDC1s. cDC1-based treatment in adjuvant and neoadjuvant contexts prevented relapse better than anti-PD-1 therapy. Neoadjuvant cDC1-based therapy boosted tumor-infiltrating CD8+ and CD4+ T resident memory (Trm) cell numbers just prior to tumor remission. Greater CD4+ Trm-like cell abundance correlated with cDC1 presence in human tumors, and patient survival.

Contributed by Paula Hochman

Succinate-loaded tumor cell-derived microparticles reprogram tumor-associated macrophage metabolism

To reprogram M2-like macrophages in the TME and enhance their antitumor efficacy, Lu and Li et al. used electroporation to load tumor cell membrane-derived microparticles with succinate (SMPs), which delivered succinate intracellularly into mitochondria and the nucleus of macrophages. SMPs, but not free succinate, induced succinylation of isocitrate dehydrogenase 2 (promoting HIF-1α stabilization) and histone H3K122 (promoting lactate dehydrogenase A expression), resulting in increased glycolysis, TCA cycle attenuation, and metabolic reprogramming of TAMs into M1 polarized macrophages. SMPs inhibited tumor progression and increased survival in several murine tumor models.

Contributed by Katherine Turner

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.