Weekly Digests
‹ Back to November

PD-1 inhibition: it’s not just for antibodies anymore

November 1, 2017

As anti-PD-1 antibody therapy can be costly and carries a risk of an autoimmune reaction, Taylor et al. explored the possibility of using a glycogen synthase kinase GSK-3 inhibitor (GSK-3i) as a small molecule inhibitor to downregulate PD-1 in the treatment of cancer.

In a recently published paper in Cancer Research, the team began with a study of metastatic tumors: they intravenously injected C57/BL6 mice with B16 melanoma tumor cells, and then treated the animals with either GSK-3i (SB415286) or anti-PD-1 every 2 days. After 14 days, the researchers analyzed the metastases in the lungs, and found that GSK-3i and anti-PD-1 showed similar levels of tumor inhibition, while combining the two therapies provided no further benefit. Similar results were observed in tumors that were allowed to establish for 7 days prior to treatment. To test for effects in primary solid tumors, the researchers intradermally injected mice with B16 cells, and then treated the animals with either GSK-3i, anti-PD-1, or GSK-3i and anti-PD-1 via intraperitoneal injections. All treatments increased survival to a similar degree.

Taylor et al. also assessed the effect of GSK-3i in an EL4 lymphoma model. EL4 cells pretreated with various concentrations of the SIINFEKL peptide of OVA were injected into the right flank, while cells that had not been pretreated were injected into the left flank of OT-1 Tg mice. Some of the mice were then treated with GSK-3i beginning on day 0 via a peritoneal injection. The team found that the growth of OVA peptide-pulsed tumor was reduced, but still visible in all animals not treated with GSK-3i. In contrast, administration of GSK-3i completely cleared the peptide-pulsed tumor mass in >80% of mice in 4 out of 7 experiments. The team also compared the efficacy of GSK-3i with anti-PD-1 and combination therapy, and found similar results across the board. Notably, at the highest tested OVA concentration (10 μg), all three treatments completely protected mice against death. In addition, similar effects on tumor growth were observed with anti-PD-L1 therapy.

The team also explored the effects of GSK-3i vs. anti-PD-1 on EL4 tumors in the absence of the OVA peptide. They found that tumors completely regressed in some mice treated with either anti-PD-1 or combination therapy, but not GSK-3i alone. Meanwhile, combination therapy improved survival over either individual therapy.

To understand how GSK-3i exerts its effect on the tumors, Taylor et al. utilized quantitative RT-PCR and flow cytometry to study its mechanism of action. They found that GSK-3i reduced pdcd1 (PD-1) and increased Tbx21 (Tbet) transcription in CD8+ T cells, and increased the percentage of tumor-infiltrating CD8+ T that expressed CD107a and granzyme B. No PD-1 downregulation was observed on CD4+ T cells or Treg cells. In genetic studies, tumor growth reduction in GSK3α/β-/- knockout mice was similar to Pdcd1-/- mice, and the addition of anti-PD-1 to GSK3α/β-/- mice or GSK-3i to Pdcd1-/- mice resulted in no further tumor reduction. GSK-3i had no direct effect on the tumor. Altogether, the data indicates that GSK-3i limits tumor growth by downregulating PD-1 expression on CD8+ T cells.

Given this proposed mechanism of action, the team explored the effect of GSK-3i in an adoptive transfer setting. Mice were intradermally injected with EL4-OVA cells, and 7 days later they underwent adoptive transfer with OT-1 cytotoxic T cells that had been cultured in vitro for 7 days in the presence or absence of OVA peptide plus GSK-3i, anti-PD-1, or a combination of the two. T cells treated only with OVA delayed the onset of tumor growth at all peptide concentrations. Pretreatment with either GSK-3i, anti-PD-1, or a combination of the two further slowed tumor growth at all peptide concentrations, and completely cleared the tumor at the highest peptide concentration. The downregulation of PD-1 expression by a single dose of GSK-3i persisted for 7-10 days.

To find out whether GSK-3i has a similar effect on human cells, Taylor et al. isolated human CD3+CD8+ T cells from peripheral blood and demonstrated that GSK-3i also downregulates PD-1 expression to a similar extent in this cell population.

The cumulative results of these experiments demonstrate that the GSK-3i effect on tumor growth is mediated by its effect on the downregulation of PD-1 and upregulation of Tbet expression in CD8+ T cells, and its efficacy is similar to anti-PD-1 therapy, indicating that GSK-3 inhibitors, which have been clinically tested in type II diabetes and neurological disorders, could serve as a potential small molecule alternative to anti-PD-1 antibody in cancer therapy.

by Anna Scherer

References:

Taylor A., Rothstein D., Rudd C.E. Small molecule inhibition of PD-1 transcription is an effective alternative to antibody blockade in cancer therapy. Cancer Res. 2017 Oct 20.

In the Spotlight...

Targeting immunosuppressive adenosine in cancer

Vijayan et al. comprehensively review the complex immunosuppressive effects of extracellular adenosine and the adenosinergic pathways on the multiple cellular and structural components of the tumor microenvironment, and their role in the development of primary and metastatic tumors. Current Phase I clinical trials, considerations for clinical development, and strategies for future therapies are also discussed.

AML-specific cytotoxic antibodies in patients with durable graft versus leukemia responses

Gillisen et al. isolated tumor-specific antibodies from B cells of acute myeloid leukemia (AML) patients who had experienced durable remission following allogeneic hematopoietic stem cell transplantation. Several antibodies recognized the U5 snRNP200 complex, which is only expressed in the nucleus and cytoplasm of healthy cells, but is expressed on the surface of AML cells. These donor-derived antibodies induced non-apoptotic cell death of AML cells in vitro, and delayed tumor growth in a SCID mouse model of AML.

A synthetic CD8alpha:MyD88 co-receptor enhances CD8+ T cell responses to weakly immunogenic and lowly expressed tumor antigens

Melanoma-specific murine CD8+ T cells engineered to mimic TLR stimulation by expressing a CD8α:MyD88 fusion protein had a lower threshold of T cell activation and improved the control of weakly immunogenic melanoma in vivo in a TCR-dependent fashion by enhancing production of IFNγ, TNFα, and co-stimulatory molecules, reducing exhaustion markers, and shifting the tumor microenvironment to a state more favorable to anti-tumor response.

Prostate cancer cells express more androgen receptor (AR) following androgen deprivation, improving recognition by AR-specific T cells

Capitalizing on the overexpression of the androgen receptor (AR) – a known mechanism of resistance to androgen deprivation therapy (ADT) – to increase the recognition of prostate cancer cells by AR-specific CD8+ T cells, Olson et al. combined ADT with an AR-targeting DNA vaccine. This combination delayed tumor growth and recurrence of castrate-resistant tumors in mouse models, providing rationale for a Phase I clinical trial, which is ongoing.

Synthetic RNA-Based Immunomodulatory Gene Circuits for Cancer Immunotherapy

Nissim et al. developed a gene circuit platform in which synthetic, cancer-specific promoters, when mutually activated, generate multiple coding RNAs that yield immunomodulatory outputs. When the platform was designed to output a combination of a cell-surface antigen (STE), a cytokine (IL-12), a chemokine (CCL21), and a checkpoint inhibitor antibody (anti-PD-1), the researchers observed T cell-mediated killing of cancer cells without harm to normal cells in vitro, and tumor reduction and prolonged survival in vivo.

A CD200R-CD28 fusion protein appropriates an inhibitory signal to enhance T cell function and therapy of murine leukemia

Oda et al. engineered immunomodulatory fusion proteins (IFPs) that link the inhibitory CD200R and stimulatory CD28 molecules to convert binding of the CD200 ligand from an inhibitory to a cell-intrinsic activating signal. Human and murine T cells expressing these IFPs showed enhanced proliferation, accumulation, and effector function in vitro. In mouse models, IFP-transduced T cells showed improved antitumor efficacy against CD200+ leukemia and did not require administration of IL-2.

Specifically differentiated T cell subset promotes tumor immunity over fatal immunity

Adoptive transfer of allogeneic T cells can lead to elimination of leukemic cells via graft-versus-leukemia reactivity, however, these cells may also cause potentially deadly graft-versus-host disease (GVHD). Ramadan et al. found that human or murine T9 cells (CD4+ and CD8+ cells differentiated under IL-4 and TGFβ) activated with IL-33 upregulate mST2, IL-9, PU.1, and amphiregulin expression, significantly reducing the severity of GVHD in mice. At the same time, T9IL-33 cells show enhanced CD8α expression for better killing of leukemia.

Effective antitumor peptide vaccines can induce severe autoimmune pathology

Using a CMV-epitope-expressing transplanted tumor in transgenic immune tolerant mice that express CMV epitopes in pancreatic islet cells, Sultan et al. show that a highly effective CMV-based vaccine can overcome tolerance and induce potent anti-tumor activity but also induce severe diabetogenic symptoms. Boosting immunization with a agonist-CD40 antibody generated T cells sensitive to negative signaling via the PD-1 axis, which was not observed when immunization was boosted with long-lived IL-2.

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.