Weekly Digests
‹ Back to September

Dream team: TGFβ and PD-L1 targeting aligns with radiotherapy to create a beneficial immune environment

September 15, 2021

Since radiotherapy (RT) can boost immune priming, it can be a great tool to convert immune “cold” tumors into “hot” immune environments, making way for effective checkpoint inhibition therapies. However, RT may also remodel the tumor microenvironment (TME), creating barriers to immune infiltration and antitumor effects. Given that one of the main contributors to TME remodeling is the TGFβ pathway, combining RT with neutralization of TGFβ and checkpoint blockade may effectively treat cold tumors. Lan, Moustafa, Knoll, and Xu, et al. investigated whether the addition of bintrafusp alfa (BA), a bifunctional fusion protein combining the extracellular domain of the TGFβRII (functioning as a TGFβ “trap”) and an anti-PD-L1 antibody, to RT (BART) can increase therapy efficacy. Their findings were recently published in Cancer Cell.

The researchers evaluated their therapeutic strategy in multiple mouse models (KPC, LLC, GL261, 4T1) that lack TIL and present with features of fibrosis, inflamed TME, and generally have limited responses to checkpoint blockade and/or radiation. In these models, BA and RT monotherapy reduced tumor burden, reduced tumor growth, and increased tumor regression. BART significantly increased these effects, resulting in prolonged survival in some of the models.

To determine what factors in the TME were affected by BART, resulting in the enhanced antitumor effects, the researchers studied fibrosis in the TME in the EMT-6 model. BA reduced α-smooth muscle actin and collagen deposition, indicative of reduced cancer-associated fibroblast (CAF) activation, and increased CD8+ TIL infiltration and repertoire. RT, on the other hand, increased activation of CAFs and deposition of collagen, which could be reversed by BART. Angiogenesis in the TME was reduced by BA, an effect that was enhanced when combined with RT.

Looking at the immune infiltrate in treated tumors, BART increased the intratumoral density of CD8+ T cells, dendritic cells (DCs), and activated NK cells, while it decreased the density of myeloid-derived suppressor cells (MDSCs). Showing the essential role of the immune infiltrate on the therapeutic effects of BART, antibody depletion of CD8+ T cells or NK cells resulted in a reduction of the inhibition of tumor growth and reduced survival.

The researchers then investigated gene expression changes induced by the therapy in the 4T1 model using RNAseq and curated gene expression signatures. Both BA and BART decreased TGFβ signature scores, and BA also decreased predictive scores of RT-induced fibrosis, extracellular matrix, epithelial-to-mesenchymal transition (EMT), vascular endothelial growth factor (VEGF) pathways, and hypoxia signatures. On the other hand, RT alone increased EMT and hypoxia signatures, while BART reversed these effects. Immune signatures also changed, with BA increasing the stimulator of interferon genes (STING) and slightly increasing IFN-associated signatures, while RT and BART upregulated these signatures even more, with RT resulting in the highest expression. Tumor-associated macrophage signatures were increased after RT, decreased after BA, and partially increased after BART. Additionally, BART increased NK cell signatures, and both BA and BART decreased MDSC signature scores, with the most prominent effect caused by BART.

The researchers then took the signatures of BART-regulated genes in the mouse model to analyze its prognostic significance for patients. Patient data from a Phase II trial using atezolizumab (anti-PD-L1) in patients with metastatic urothelial cancer were clustered based on the discovered BART-regulated genes. This resulted in three patient groups, which showed variable survival outcomes, in contrast to clustering based on immune signatures or monotherapy-related genes. Patients in cluster 3 had improved outcomes, reduced expression of TGFβ signaling-related genes, and increased expression of immune activation genes, suggesting prognostic significance of this gene signature.

Studying the immune effects of BART further, the 4T1 model was used to assess whether the treatment could enhance antitumor responses and affect metastases. Tumor-bearing mice with lung metastases were treated systemically with BA, while the primary tumor received RT, using a small animal radiation research platform that mimics localized RT in patients. While the monotherapies reduced the tumor burden of the primary tumors, BART reduced both primary and lung tumor burden, more so than BA alone, suggesting a systemic effect.

Given that these systemic effects might be caused by the induction of antitumor T cell responses, the researchers assessed the immune infiltrates in metastatic lesions. The density of CD8+ T cells and the ratio of CD8+ T cells to Tregs increased after BA and BART, with the most significant increase after BART. When CD8+ T cells or NK cells were depleted, the inhibition of primary tumor growth induced by BART was partially abrogated. Additionally, depletion of CD8+ T cells abrogated the BART-induced tumor reduction effects on metastases, suggesting the essential role of CD8+ T cells in the abscopal effects of BART.

The 4T1 lung metastasis model is characterized by pulmonary myeloid cell infiltration, but this was reduced after BA and, more significantly, BART. BART decreased the total number of polymorphonuclear (PMN)-MDSCs, in contrast to monotherapies. Additionally, RT and BART reduced the number of monocytic (M)-MDSCs.

To investigate the effects of BA on RT-induced lung fibrosis, mice were treated with RT, resulting in increased lung density and reduced lung volume size, indicative of tissue remodeling. The addition of BA treatment resulted in a lower lung density and protected the lung volume, while anti-PD-L1 or a TGFβ trap alone did not affect it. In addition, post-RT BA reduced lung fibrosis, preserved lung tissue architecture, decreased collagen deposition, normalized alveoli, and protected pulmonary function. Using single-cell RNAseq, mouse lungs treated with RT or BART were subjected to tSNE plotting of cellular populations. Fibrosis-associated markers were then used to identify cell populations critical for the lung fibrosis seen after RT. PD-L1 was mainly expressed at high levels in lipofibroblast-like clusters and in some endothelial clusters, which also expressed TGFβ. These data suggest that BA may target the TGFβ trap to relevant populations that also express PD-L1.

Together, these data show that radiotherapy and co-targeting TGFβ and PD-L1 can stimulate antitumor immune responses and overcome immune barriers in immunologically cold tumors, creating an effective “in situ vaccine”, while simultaneously overcoming some of the adverse fibrotic effects caused by RT in the TME. These data support the clinical translation of this approach.

Write-up by Maartje Wouters, image by Lauren Hitchings

Meet the researcher

This week, senior author Amir Abdollahi answered our questions. 

From left to right, top panel: Yan Lan, Mahmoud Moustafa, Maximilian (Max) Knoll, Jennifer Furkel, Chunxiao Xu; low panel: Tsz-Lun Yeung (So)​, Molly Jenkins, Katrin Rein, Kin-Ming Lo, Amir Abdollahi

What prompted you to tackle this research question?​
Despite the rapid progress of immune oncology (IO), unfortunately, still a large fraction of cancer patients do not benefit from immunotherapy. In certain entities such as primary brain tumors (glioblastoma) and pancreatic cancer, the prognosis remains poor, despite evaluation of several treatment escalation strategies. Our motivation was therefore to discover a potentially universal strategy to reconstitute tumor immune surveillance to cure these therapy-resistant tumors. Modern high-precision radiotherapy (RT) delivers high-energy beams to individualized tumor volumes identified by modern non-invasive imaging techniques (CT-scan, MRI , PET). RT elicits direct antitumoral, anti-angiogenic and immunogenic effects.Therefore, potent activation of the immune system could be confined to the irradiated tumor volume. However, these positive effects of RT are, in part, counterbalanced by immune evasion and tissue remodeling processes, limiting exploitation of its full therapeutic potential. Activation of immune checkpoints (e.g., PD-L1) and expression of TGFβ are among the most relevant undesired RT effects detected at the tumor microenvironment (TME) interface. We hypothesized that reprogramming the TME via trapping of TGFβ combined with anti-PD-L1 immune checkpoint blockade may synergize with radiotherapy towards development of an effective in situ tumor vaccination strategy. In fact, we could show that immune activation by RT and TME reprogramming via simultaneous targeting of PD-L1/TGFβ (Bintrafuspalpha, BA) are together required for induction of a CD8+ T cell-dependent tumor-specific immunity. In line, BART combination not only improved tumor responses at the irradiated tumor sites, but also led to regression of abscopal metastases, outside the irradiation field.

What was the most surprising finding of this study for you?
Perhaps the most surprising finding was that BA, but not TGFβ trap alone was able to attenuate undesired late radiation-induced normal tissue toxicity (lung fibrosis). Single-cell analysis revealed that key TGFβ-expressing subpopulations (i.e. clusters of endothelial cells and M2-like lipofibroblasts) also expressed PD-L1. Therefore, in addition to combining two IO modalities, the anti-PD-L1 arm of BA may target TGFβ trap to the relevant populations. This discovery may now open the field for development of next generation IO drugs with enhanced efficacy and possibly reduced toxicity by selective targeting of therapeutic modules to specific cellular compartments, rather than global inhibition or activation of signaling cascades.

What was the coolest thing you’ve learned (about) recently outside of work? 
Among a plethora of lessons we all learned from the COVID-19 pandemic, time-lapse tracking of SARS-COV2 evolution was/is intriguing and instructive. It started with the appearance of the D614G mutation in Europe that would later dominate the global pandemic. It continued with the formation of the N501Y variant in UK/alpha and almost simultaneously the beta/gamma variants in South Africa and Brazil with additional mutations in 484 and 417 positions of the RBD. The community was preparing for these converging mutations and anticipated a narrow evolutionary landscape for viral escape. Within only a couple of months, the world is now dominated by the delta variant 617.2 that started its rise in India and lacks all these mutations. The delta variant, rather, combines the two mutations previously found in California (L452R) and Mexico (T478K). Time will show if the predicted convergence of immune-escape mutations will resurrect once the global immunization rate increases, or novel residues or completely new variants will emerge. The challenge in prediction of the evolutionary landscape of a relatively genetically stable coronavirus system teaches us that we need to substantially gear up our knowledge to understand or even predict the evolution of the tremendously more complex and genetically unstable tumor model system. 

References:

Lan Y., Moustafa M., Knoll M., Xu C., Furkel J., Lazorchak A., Yeung T.L., Hasheminasab S.M., Jenkins M.H., Meister S., Yu H., Schlegel J., Marelli B., Tang Z., Qin G., Klein C., Qi J., Zhou C., Locke G., Krunic D., Derner M.G., Schwager C., Fontana R.E., Kriegsmann K., Jiang F., Rein K., Kriegsmann M., Debus J., Lo K.M., Abdollahi A. Simultaneous targeting of TGF-β/PD-L1 synergizes with radiotherapy by reprogramming the tumor microenvironment to overcome immune evasion. Cancer Cell. 2021 Sep 7.

In the Spotlight...

Tumour-targeted interleukin-12 and entinostat combination therapy improves cancer survival by reprogramming the tumour immune cell landscape

Using 3 mouse tumor models, Hicks et al. tested a combination therapy consisting of entinostat (a histone deacetylase inhibitor that increases tumor necrosis) and IL-12 (targeted to the TME via coupling to NHS76, an antibody that binds exposed DNA in necrotic areas). Therapy was well tolerated and resulted in increased tumor necrosis, TME accumulation of IL-12, eradication of established tumors, tumor-specific resistance to rechallenge, increased chemokine-driven T cell infiltration, sustained CD8 TIL activation and function, decreased Tregs, TME polarization towards inflammation, and promotion of M1-like (over M2- like immunosuppressive) TAMs.

Contributed by Margot O’Toole

Low Dose Radiotherapy Reverses Tumor Immune Desertification and Resistance to Immunotherapy

Herrera et al. demonstrated that low-dose radiotherapy (LDRT) can be used on immune-cold metastatic tumors in high volumes without toxicity, and reprograms the TME to promote immune cell infiltration and responses to immunotherapy. LDRT sensitized tumors to a rationally designed immunotherapy combination in a CD4+ and CD8+ T cell-dependent manner, expanded T cells with progenitor exhausted and exhausted features (with a subset expressing NKG2D), and mobilized activated dendritic cells expressing the NKG2D ligand Rae1. In phase I clinical trial, combinatorial treatment regressed metastatic solid tumors and increased T cell influx in responding patients.

Contributed by Shishir Pant

A modular self-adjuvanting cancer vaccine combined with an oncolytic vaccine induces potent antitumor immunity

Taking advantage of the antigen (Ag)-specific T cell induction of a protein vaccine (containing a cell-penetrating peptide, Ag, and a peptide TLR4 agonist) and the powerful innate-adaptive stimulation by a VSV oncolytic virus (OV) carrying the same Ag, Das and Belnoue et al. demonstrated that s.c. protein vaccine priming followed by i.v. OV boosting led to the optimal Ag-specific CD8+ T cell responses (number and quality), resulting in significant TME remodeling and tumor control in multiple models. Resistance to complete tumor control was associated with the presence of early exhausted T cells, and addition of anti-PD-1 axis therapy led to more CRs.

Contributed by Ed Fritsch

A HER2 Tri-Specific NK Cell Engager Mediates Efficient Targeting of Human Ovarian Cancer

Vallera et al. bioengineered CAM1615HER2, a tri-specific killer engager (TriKE) consisting of an antibody fragment recognizing CD16 and a scFv recognizing HER2, linked by a human IL-15 molecule. In vitro, CAM1615HER2 enhanced NK cell expansion; increased degranulation and IFNγ production in response to HER2+ target cells; and increased NK cell-mediated killing of a variety of HER2+ cancer cell lines and ovarian cancer spheroids. When using NK cells taken from patient tumors (vs. healthy donor PBMCs), these effects were less pronounced. In a xenograft mouse model, CAM1615HER2 mediated tumor control and increased survival.

Contributed by Lauren Hitchings

Integrin-α (V)-mediated activation of TGF-β regulates anti-tumour CD8 T cell immunity and response to PD-1 blockade

Using tumor specimens from patients with NSCLC, Malenica et al. showed that lower αv levels were associated with better anti-PD-1 axis-related PFS and increased density of CD103+CD8+ TRM TILs. Ex vivo, tumor-expressed αv integrins contributed to activation of TGF-β to induce CD103 expression on activated CD8+ T cells. In mouse modeling, αv ablation from tumor cells increased activated PD-1+Ki-67+ KLRG1+CD8+ T cells and correlated with decreased active TGFβ intratumorally. PD-1 blockade improved control of αv knockout tumors and increased granzyme B+CD69+CD103+CD8+ TRM TILs; this was inhibited by intratumoral CD103 blockade.

Contributed by Paula Hochman

Functional HPV-specific PD-1+ stem-like CD8 T cells in head and neck cancer

Eberhardt and Kissick et al. characterized CD8+ T cells in patients with human papillomavirus (HPV)-positive HNSCC and identified a substantial proportion of HPV E2-, E5- and E6-specific CD8+ T cells in tumors, but not peripheral blood. Single-cell sequencing revealed 3 transcriptionally distinct subsets: stem-like cells expressing high levels of TCF7 and other stemness-associated genes; transitory cells expressing effector molecules; and terminally differentiated cells expressing high levels of TIM-3 and effector molecules. These subsets shared TCR clonotypes, and pseudotime analysis suggested the progression from stem-like to transitory to terminally differentiated states.

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.