Weekly Digests
‹ Back to August

The timing of priming and PD-1 blockade

August 7, 2019

Checkpoint blockade of the PD-1 axis (PD-1 or PD-L1) can be effective in inducing antitumor immunity, although many patients still relapse or fail to derive clinical benefit at all. Combinations of anti-PD-1 treatment with vaccines against tumor-associated antigens or with other immunomodulatory agents are actively being explored, and optimal timing and sequencing have yet to be established. In clinical trials combining a vaccine with anti-PD-1, anti-PD-1 treatments often begin earlier for logistical reasons. Recently, Verma et al. investigated whether the sequencing of anti-PD-1 and vaccine treatments might influence therapeutic outcomes; their results were published in Nature Immunology.

Investigating the common clinical sequence in which anti-PD-1 treatment is delivered early followed later by concomitant treatment with anti-PD-1 and vaccine, Verma et al. implemented this schedule in mice bearing TC-1 lung cancer or B16 melanoma (both resistant to anti-PD-1 and vaccines as monotherapies) and saw no significant therapeutic benefit. When no anti-PD-1 pre-treatment was given, however, coadministration of anti-PD-1 and vaccine induced a synergistic antitumor effect that slowed tumor growth. Profiling of T cell infiltrates from tumors showed that vaccine alone induced tumor antigen-specific T cells, and that this effect was even more significant when anti-PD-1 was coadministered with the vaccine. When anti-PD-1 pretreatment was administered, however, antigen-specific T cells decreased in response to subsequent vaccine or vaccine/anti-PD-1 combination therapy.

To better understand why anti-PD-1 pre-treatment hindered later therapies, Verma et al. examined overall and antigen-specific CD8+ T cell populations and identified signs of early and ongoing apoptosis in both. The researchers determined that anti-PD-1 did not enhance the activation of CD8+ T cells, and reasoned that apoptosis was not likely to have been caused by activation-induced cell death. Considering that anti-PD-1 could be causing cell death by altering the activation process, the researchers investigated the effects of anti-PD-1 and antigen stimulation in gp100-specific CD8+ T cells. When anti-PD-1 was administered in combination with gp100 peptide (to stimulate the TCR), TCR signaling was fully functional and cells took on an activated (CD40L+) effector (IFNγ-producing) phenotype. When anti-PD-1 was administered before gp100, however, it altered the phosphorylation patterns involved in TCR signaling, thereby interfering with TCR signaling in response to antigen stimulation. This drove CD8+ T cells towards a dysfunctional state, and ultimately towards their death.

PD-1+CD38hi CD8+ T cells are a subset of T cells that have been previously described as being dysfunctional and unresponsive to antigen restimulation, and Verma et al. found that pre-treatment with anti-PD-1 significantly increased overall CD38 expression, as well as the portion of PD-1+CD38+ T cells within both total CD8+ and antigen-specific CD8+ T cell populations. Vaccine treatment alone induced no change in CD38 expression, while coadministration of vaccine with anti-PD-1 decreased CD38 and the portion of PD-1+CD38+ cells within total CD8+ and antigen-specific CD8+ T cell populations. PD-1+CD38+ CD8+ T cells induced by anti-PD-1 pretreatment were dysfunctional (they did not show signs of activation upon antigen restimulation) and underwent apoptosis at an increased rate. Likely due to the increased rate of apoptosis within CD8+ T cells, mice pre-treated with anti-PD-1 also generated smaller fractions of central and effector memory CD8+ T cells.

To determine whether the PD-1+CD38+ CD8+ T cells that arose following anti-PD-1 pre-treatment were to blame for the failure of subsequent combination therapy, Verma et al. activated gp100-specific T cells in vitro in the presence of anti-PD-1 and adoptively transferred the total cultures or cultures depleted of PD-1+CD38+ cells (about 30% of the total culture) into mice. The researchers found that the PD-1+CD38+-depleted culture more effectively reduced tumor growth and increased survival. When expression of CD38 was genetically knocked down in cultures of dysfunctional PD-1+CD38+ CD8+ T cells, the cells regained their ability to proliferate, to become activated, and to express effector molecules, linking CD38 directly to dysfunctionality.

Given that anti-PD-1 pretreatment induced dysfunctional PD-1+CD38+ CD8+ T cells, while anti-PD-1 in combination with vaccine did not, Verma et al. investigated whether the quality of T cell priming may play a role. Using OVA-specific T cells in vitro, Verma et al. compared the impact of anti-PD-1 in the context of optimal priming (stimulation with high-affinity OVA) or suboptimal priming (no stimulation, or stimulation with low-affinity OVA-V). While blocking PD-1 under optimal priming conditions had no impact on PD-1+CD38+ CD8+ T cell production, blocking PD-1 under suboptimal priming conditions or before optimal priming drove T cells towards a PD-1+CD38+ phenotype and apoptosis-mediated cell death. Similar results were observed in tumor-bearing mouse models of suboptimal and optimal priming.

To determine the relevance of PD-1+CD38+ CD8+ T cells in human cancers, Verma et al. evaluated immune infiltrates from tumor biopsies from patients with metastatic melanoma taken before or after anti-PD-1 or combination anti-PD-1/anti-CTLA-4 therapy. They found that before and after checkpoint blockade, non-responding lesions contained a higher fraction of PD-1+CD38+ CD8+ T cells compared to responding lesions, suggesting that the fraction of PD-1+CD38+ cells within infiltrating CD8+ T cells in the tumor could serve as a biomarker of response. Further, most responders showed a decrease in the fraction of CD38+ cells within PD-1+CD8+ T cells at nine weeks post-therapy, while non-responders showed stabilization or an increase in CD38+ cells, which could serve as an easy-to-access biomarker of on-treatment response.

Overall, Verma et al. show that administration of PD-1 blockade under suboptimal priming conditions drives the production of a PD-1+CD38hi CD8+ T cell population that interferes with antitumor immunity and prevents therapeutic responses. This evidence may help to explain why many cancer patients fail to derive clinical benefit from checkpoint blockade and combinations, as well as why patients with a higher mutation burden (i.e. more opportunity for optimal on-site priming) are more likely to respond. Improving the antigenicity of tumors or correctly sequencing PD-1 blockade and vaccines in current and future clinical trials could serve as important strategies in improving antitumor responses.

by Lauren Hitchings

Meet the researcher

This week, lead author Samir Khleif answers our questions.

Lead author Samir Khleif

What prompted you to tackle this research question?
As a cancer immunologist and a physician who treats patients, it was wonderful to see anti-PD-1 therapy work for so many patients. However, at the same time it was painful to see many patients not getting the benefits out of their anti-PD-1 therapy. This had us thinking about the reasons that lead to the non-response in these patients. To address this issue, we turned our attention to model systems that could be employed to answer the question regarding resistance to anti-PD-1 therapy in humans. Accordingly, we made some observations where we saw that initial activation status of the killer lymphocytes had a great impact on the therapeutic outcomes seen after anti-PD-1 therapy. Steadily building upon our initial observations, we discovered this new mechanism of resistance to anti-PD-1 therapy wherein we found that a single dysfunctional cell type can be responsible for therapeutic failures.

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

The most surprising finding of this study was the observation that the very treatment agent by itself can turn rogue and contribute to failure of the therapy. Though, of course, other contributing factors, such as suboptimal priming conditions, should be there to make the environment conducive for generation of dysfunctional cells that render the treatment ineffective.

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

I recently visited the Amazon and, amongst the many things that I learned about the eco harmony and animal behavior is the beautiful synchronized musical symphony that cicadas make during the mating season. This was discovered by scientists using sound-slowing techniques. Besides the amazing musical tune that sounds like a Middle-Age Viennese Choir, it tells a lot about nature organization and lack of randomness in biology. It turned out that other species do the same, including crickets.

References:

Verma V., Shrimali R.K., Ahmad S., Dai W., Wang H., Lu S., Nandre R., Gaur P., Lopez J., Sade-Feldman M., Yizhak K., Bjorgaard S.L., Flaherty K.T., Wargo J.A., Boland G.M., Sullivan R.J., Getz G., Hammond S.A., Tan M., Qi J., Wong P., Merghoub T., Wolchok J., Hacohen N., Janik J.E., Mkrtichyan M., Gupta S., Khleif S.N. PD-1 blockade in subprimed CD8 cells induces dysfunctional PD-1(+)CD38(hi) cells and anti-PD-1 resistance. Nat Immunol. 2019 Jul 29.

In the Spotlight...

Clonal replacement of tumor-specific T cells following PD-1 blockade

Single-cell RNAseq and TCRseq on site-matched biopsies from patients with basal or squamous cell carcinoma before and after anti-PD-1 treatment revealed that post-treatment clonally expanded TILs were enriched in tumor-specific, exhausted CD8+ T cells, many of which exhibited TCR clonotypes that were not detected in the tumors prior to treatment and appeared to represent new antigen specificities. These results suggest that pre-existing exhausted TILs have limited capacity for reinvigoration and are replaced by newly recruited cells which themselves become exhausted.

CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy

Barkal et al. showed that CD24, a GPI-linked protein overexpressed on ovarian, TNBC, and other solid tumors, inhibited phagocytosis through interaction with Siglec-10 on tumor-associated macrophages. Siglec-10+ macrophages cultured with MCF-7 breast cancer cells and anti-CD24 exhibited potentiated phagocytosis that was synergistic with anti-CD47. In patients, low CD24 levels correlated with increased relapse-free and OS in ovarian and breast cancer. In mice, treatment of MCF-7 tumors with anti-CD24 significantly decreased tumor burden, suggesting the potential of CD24 blockade for immunotherapy.

Contributed by Katherine Turner

Assessment of TCR signal strength of antigen-specific memory CD8+ T cells in human blood

Wu et al. used flow cytometry of ex vivo stimulated human blood cells to show that all activated CD8+ T cell subsets upregulate interferon regulatory factor 4 (IRF4) expression proportionate with TCR, not cytokine, signal strength. Antigen-stimulated T cell expression of IRF4 and CD137 (4-1BB) enabled sensitive detection of antigen-specific CD8+ T cells. Antigen-stimulated HIV-specific CD8+ T cells from chronic HIV+ subjects revealed an IRF4loCD137loCD25lo population and increased CD27 (memory cell marker) expression, suggesting that suboptimal TCR signaling and high memory cell frequency underlie impaired CD8+ T cell function.

Contributed by Paula Hochman

In situ vaccination with defined factors overcomes T cell exhaustion in distant tumors

Intratumor delivery of TLR4 agonist monophosphoryl lipid A and agonistic anti-CD40 in combination with anti-PD-1 (CMP) regressed treated and distant matched tumors in multiple bilateral murine tumor models. CMP activated CD11chi cells in tumors, which were later found in the draining lymph nodes (LNs). CMP boosted proliferation of both endogenous and adoptively transferred tumor antigen-specific CD8+ T cells and reduced exhausted CD8+ T cells in treated and distant tumors, but not in the spleen and LNs. Melanoma patients with pre-treatment tumor gene sets matching the CMP-induced signature had improved progression-free survival on nivolumab.

Contributed by Alex Najibi

CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity

To prevent glioblastoma (GBM) escape via antigen loss, Choi et al. designed CART.BiTE cells that express a CAR specific for EGFRvIII (a tumor-specific antigen expressed in many GBM tumors) and secrete a bispecific T cell engager against EGFR (overexpressed in GBM, but also expressed in normal tissues). Intracranially-delivered CART.BiTE cells cleared heterogeneous GBM tumors in mice. BiTEs redirected both CAR T cells and bystander T cells to tumor cells, and activation via CAR and BiTE together led to a less exhausted TCM phenotype. CART.BiTE cells did not cause toxicities in a human skin graft model.

CLINICAL TRIAL: A Randomized Double-Blind Placebo-Controlled Phase 2 Trial Of Dendritic Cell Vaccine ICT-107 In Newly Diagnosed Patients With Glioblastoma

In a double-blind phase II clinical trial, 124 HLA-A1+ and/or -A2+ patients with newly-diagnosed, resected glioblastoma were randomized 1:2 to intradermally receive placebo or ICT-107, a vaccine consisting of autologous DCs pulsed with six epitopes (MAGE-1, HER-2, AIM-2, TRP-2, gp100, IL-13Rα2). The vaccine was well tolerated. ICT-107 statistically significantly increased PFS by 2.2 months, with maintenance of quality of life, but did not increase OS. HLA-A2+ patients had higher tumor antigen expression and higher immune response than HLA-A1+ patients, leading to longer survival.

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.