Weekly Digests
‹ Back to July

Radiotherapy from the inside out? TRT enhances immunotherapy

July 28, 2021

In the setting of cancer, radiotherapy can directly damage or kill tumor cells, and can act as an in situ vaccine that triggers antitumor immunity; the contribution of each of these mechanisms varies based on the dose. In most cases, radiation is delivered as targeted external beam radiotherapy (EBRT), but not all tumors and metastases are accessible for treatment. Patel et al. recently investigated the possibility of delivering low-dose radiotherapy through targeted radionuclide therapy (TRT). This strategy utilized an alkylphosphocholine analog, NM600, which is robustly taken up by a variety of mammalian tumors, to carry and deliver radioactive yttrium (90Y). 90Y-NM600 was tested in a variety of models and in combination with a variety of therapies. The results were recently published in Science Translational Medicine.

Patel et al. began by using PET imaging, pharmacokinetic biodistribution studies, and dosimetry calculations to show that 90Y-NM600, delivered intravenously into mice bearing B78 (a B16 melanoma derivative), NXS2, or 4T1 flank tumors, preferentially accumulates in tumors, especially in larger tumors, compared to in normal tissue. Using a dose that was previously shown to not reduce white blood cell or lymphocyte counts, the researchers showed 90Y-NM600 could also be safely combined with anti-CTLA-4 without significant toxicity.

Next, the researchers evaluated the potential antitumor efficacy of 90Y-NM600 in various settings. In combination with anti-CTLA-4, 90Y-NM600 improved antitumor responses and increased survival in a dose-dependent manner. The two highest doses of 90Y-NM600, 50 μCi and 100 μCi, enhanced the efficacy of CTLA-4 to a similar degree, so 50 μCi was used going forward. In addition to the dose, the researchers evaluated the effects of the timing and sequence of the dose, and found that the variations tested had little impact on the success of therapy. The addition of anti-PD-1 to this regimen also did little to improve the effects. Only mice that received 90Y-NM600 in combination with immunotherapy achieved complete responses, and responding mice rejected rechallenge with the same tumor after 90 days. Further, mice bearing 4T1 tumors, which spontaneously develop metastases, showed reduced numbers of metastatic lung nodules when treated with 90Y-NM600 and anti-CTLA-4.

A major potential advantage TRT compared to EBRT is the ability to reach and treat all sites of metastatic disease without suppressing the immune system. Comparing these different treatment strategies, Patel et al. treated tumor-bearing mice with anti-CTLA-4 plus targeted EBRT, whole-body EBRT, or TRT. Mice treated with targeted EBRT or TRT showed similar antitumor responses, but the effect was lost in mice treated with systemic EBRT, likely due to systemic immunosuppression. Consistent with this, the antitumor effects of EBRT and TRT were dependent on T cells.

Evaluating the mechanism behind the antitumor effect of TRT versus low- and moderate-dose EBRT, Patel et al. looked at the tumor immune environments in treated mice. Moderate-dose EBRT initially decreased infiltrating immune cells, but these cells rebounded at days 7 at 14. Low-dose TRT on the other hand, improved the tumor immune environment starting at day 1, with increased innate myeloid cells and natural killer cells, and an increased CD8+ T cell/Treg ratio. Looking at immune-related gene expression, the researchers also observed a number of changes that began at day 1, peaked at day 7, and were trending towards baseline by day 14. These included a modest decrease in Il6 in TRT-treated mice, an increase in Vcam1 in TRT- and low-dose EBRT-treated groups, and increased expression of Fas (indicative of apoptosis) across treatment groups. Increased IFNβ, indicative of increased activation of the cGAS/STING pathway, was also observed across all groups, though the moderate-dose EBRT group showed the greatest increases in downstream type I IFN response genes and MHC-1. Still, activation of the cGAS/STING pathway was found to be required for antitumor efficacy in TRT-treated mice as well.

Further investigating changes in the TME induced by TRT plus anti-CTLA-4, the researchers observed increased CD45+ immune cells, CD3+ T cells, CD8+ effector T cells, CD8+CD103+ tissue resident effector memory T cells, and γδ T cells. They also saw a decrease in PD-1, suggesting reduced immune exhaustion. Most cytokines also increased in the TME, particularly in responding mice. One exception was IL-10, which was reduced in the TME, consistent with a reduction of infiltrating Tregs or suppressive monocyte populations. Ex vivo, TILs isolated from combination-treated animals produced more IFNγ upon stimulation. Further, an increase in clonal expansion of T cells was also observed through deep TCRβ sequencing. Interestingly, an increase in T cell diversity was not observed, despite the fact that EBRT and immune checkpoint therapy treatment had previously been shown to increase T cell diversity.

Given that targeted moderate-dose EBRT and low-dose TRT have distinct mechanisms for eliciting antitumor efficacy, Patel et al. hypothesized that they could be combined for an additive effect. Delivered together, these two forms of radiotherapy enhanced the antitumor effect of CTLA-4 more than either individual treatment, and a strong abscopal effect was observed. Mice that experienced a complete response in both tumors (B78) were resistant to rechallenge with the same tumor line, and most were resistant to a later rechallenge with the parental B16 melanoma, which shares common antigens.

To test the safety and feasibility of 90Y-NM600 in a larger animal model that would more closely mimic human patients, Patel et al. turned to companion canines with cancer. Two dogs with metastatic cancer (osteosarcoma and melanoma) were enrolled in the study and were treated with a combination of targeted EBRT and systemic TRT. Imaging showed semi-selective uptake of 90Y-NM600 into the primary tumors and in all documented metastasis. Little evidence of toxicity was observed through laboratory testing, and no adverse events occurred. Uptake of 90Y-NM600 was also low enough in the bone marrow that the systemic radiation did not induce immunosuppression from bone marrow depletion.

Overall, Patel et al. showed that TRT, delivered in the form of 90Y-NM600, can safely modulate tumor immune environments and enhance the effectiveness of immune checkpoint blockade. Because 90Y-NM600 is selectively taken up by tumors, it can be delivered systemically and can reach tumor metastases that may not be accessible for treatment with radiotherapy through EBRT. Further, as these two forms of radiotherapy work through different mechanisms, they could be combined to further enhance the efficacy of immune checkpoint blockade, particularly with anti-CTLA-4.

Write-up and image by Lauren Hitchings

MEET THE RESEARCHER

This week, first author Ravi Patel answered our 3 questions.

What prompted you to tackle this research question?
As a radiation oncologist, I’ve always felt that we were limited in our ability to treat patients with metastatic cancer. At most, we could only offer disease palliation for these patients. When I was completing residency, I wanted to work on research in which I could make a meaningful impact in treating these patients’ disease with novel radiation approaches. Therefore, I decided to pursue a Benston Research Fellowship with Dr. Zach Morris and Dr. Paul Sondel to study combination radiation immunotherapy approaches. I was fortunate enough to fall into an early collaboration between the Morris-Sondel group and Dr. Jamey Weichert, where they were working on using targeted radiotherapy in combination with an immunocytokine (anti-GD2-IL2) developed by their group. For my project, I chose to work on a bit broader approach, testing the ability of these targeted radiotherapy agents to enhance the efficacy of standard clinical immune checkpoint inhibitors in a tumor agnostic fashion. I’m excited about our initial results and I believe this type of systemic low-dose targeted radiotherapy approach can make an impact in improving outcomes in patients with metastatic cancer.

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

The most surprising finding from our study was that such low doses of radiation were able to stimulate such a strong antitumor immune response when combined with checkpoint blockade in these immunotherapy-resistant tumor models. For example, studies in hepatocellular carcinoma have shown that radiation doses ranging from 120 to more than 200 Gy are needed to treat with Yttrium-90 monotherapy in these relative radiosensitive tumors. In contrast, our study utilizes Yttrium-90 radiation absorbed in doses of approximately 2.5 Gy to enhance the efficacy of immune checkpoint blockade. Moreover, we found that additional targeted radiopharmaceutical therapy dose escalation didn’t lead to further gains in efficacy. This finding was a surprise to us; however, it can help us greatly limit toxicity in patients when we translate this approach into human trials.

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

I’m in Pittsburgh now and loving my new city, but since I started this project in Wisconsin, I’ll mention a hobby I picked up there. As many people know, Wisconsin has many wonderful lakes, so during my research fellowship I decided to join a sailing club. I learned to sail at the same time as I started this research project and learned about cancer immunotherapy (both areas were completely new to me at the time). As a new sailor, I often found myself initially struggling, since my instinct was to fight the wind and I would find myself “In Irons” or too “close-hauled” for the conditions. However, as I gained experience, I learned to let the wind guide me and make more subtle adjustments. Looking back, the lessons I learned sailing were also applicable to my research work, as well as outside life.

References:

Patel R.B., Hernandez R., Carlson P., Grudzinski J., Bates A.M., Jagodinsky J.C., Erbe A., Marsh I.R., Arthur I., Aluicio-Sarduy E., Sriramaneni R.N., Jin W.J., Massey C., Rakhmilevich A.L., Vail D., Engle J.W., Le T., Kim K., Bednarz B., Sondel P.M., Weichert J., Morris Z.S. Low-dose targeted radionuclide therapy renders immunologically cold tumors responsive to immune checkpoint blockade. Sci Transl Med. 2021 Jul 14.

In the Spotlight...

Acquired resistance to anti-MAPK targeted therapy confers an immune-evasive tumor microenvironment and cross-resistance to immunotherapy in melanoma

Haas et al. demonstrated that resistance to targeted therapy (TT) in melanoma leads to activation of a cancer cell–intrinsic signaling program with enhanced and altered transcriptional output associated with an immune-evasive TME and cross-resistance to immunotherapy, with implications regarding scheduling of TT and immunotherapies. The immune-evasive TME was characterized by reduced infiltration and functionality of CD103+ DCs and T cells. Inhibition of the MAPK pathway in RAFi-resistant tumors increased tumor infiltration and maturation of CD103+ DCs, reduced suppressive myeloid cells, increased T cell infiltration, and improved tumor control.

Contributed by Shishir Pant

Anti-tumor effects of RTX-240: an engineered red blood cell expressing 4-1BB ligand and interleukin-15

McArdel et al. showed that red blood cell progenitors transduced in vitro to express immunomodulators, and then cultured through expansion, differentiation, and maturation phases, gave rise to a candidate drug delivery system: Red Cell Therapeutic (RCT). In vitro results showed that one such RTC (RTX-240), expressing both 4-1BBL and IL-15/IL-15Rα fusion (IL-15TP) on the surface, augmented anti-CD3-dependent activation of human CD8+ T cells and provided direct stimulation to both memory CD8+ T cells and NK cells. In two mouse models, 4-1BBL and IL-15(TX) on RBCs inhibited tumors without toxicity or unwanted off-tissue effects.

Contributed by Margot O’Toole

Neoantigen vaccination induces clinical and immunologic responses in non-small cell lung cancer patients harboring EGFR mutations

In a phase I trial, 24 late-stage NSCLC patients were immunized with personal neoantigen peptides (based on mutations detected by a 508 gene oncogene panel) and topical Imiquimod. The treatment was safe, induced objective responses in 7/16 EGFR-mutant (but 0/8 EGFR-wild-type) patients, and further extended survival with EGFR inhibitor therapy. Neoantigen-specific T cells were detected in PBMCs (predominantly in patients with EGFR-mutant NSCLC), were mainly targeting EGFR-mutant peptides, and were associated with treatment response. In an EGFR-mutant tumor cell line, EGFR inhibition increased antigen presentation gene expression and chemokine production.

Contributed by Alex Najibi

Combining CD47 blockade with trastuzumab eliminates HER2-positive breast cancer cells and overcomes trastuzumab tolerance

To address resistance that occurs in the majority of advanced HER2+ breast cancer following trastuzumab therapy, Upton et al. evaluated trastuzumab combined with macrophage checkpoint immunotherapy. Inhibition of CD47, which functions as a “don’t eat me” signal and is expressed in many malignancies, by Hu5F9-G4 (magrolimab) stimulated antibody-dependent cellular phagocytosis (ADCP) by macrophages. A trastuzumab and magrolimab combination was evaluated in trastuzumab-resistant, ADCC-tolerant Her2+ tumor models, resulting in Fc-dependent macrophage phagocytosis, superior efficacy, and increased survival.

Contributed by Katherine Turner

TIGIT-Fc Promotes Antitumor Immunity

To interrupt the interactions between the adhesin TIGIT (found on NK and T cells) and its primary ligand CD155 (PVR; a dysregulated adhesin found on many tumor cells), Shen, Fu, and Wei et al. tested human and mouse versions of a dimeric fusion of the extracellular domain of TIGIT to Ig-Fc (TIGIT-Fc). TIGIT-Fc demonstrated potent ADCC activity against multiple tumor lines and inhibition of Treg suppression in vitro. Alone and in combination with anti-PD-L1, TIGIT-Fc induced antitumor activity in xenograft models (with allogeneic human T cells) and in syngeneic tumor models. Activity was dependent on NK and CD4+ T cells, and generated immunological memory.

Contributed by Ed Fritsch

Bempegaldesleukin Plus Nivolumab in First-Line Metastatic Melanoma

Diab et al. reported the safety and clinical efficacy of CD122-preferential interleukin-2 pathway agonist bempegaldesleukin (BEMPEG) plus nivolumab in a first-line advanced disease melanoma setting. The BEMPEG plus nivolumab combination was well tolerated, and efficacy compared favorably to nivolumab alone in 38 evaluable patients. The median PFS was 30.9%, the OS at 24 month was 77%, and 47.4% of patients experienced complete clearance of target lesions. Treatment induced polyfunctional CD8+ and CD4+ T cell responses and baseline polyfunctionality. HLA-E expression and an increase in eosinophils in blood were associated with higher ORR.

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.