Weekly Digests
‹ Back to August

PD-1-high TILs: a predictive biomarker for ICB in lung cancer

August 3, 2022

Despite the promise of immune checkpoint blockade (ICB) therapy in treating non-small cell lung cancer (NSCLC), the majority of patients do not respond and toxicity remains a major issue. Identifying biomarkers of ICB efficacy could direct treatment to responsive patient populations, but remains an ongoing challenge in the cancer immunotherapy field. Now, reported in Clinical Cancer Research, Hummelink et al. pinpoint a predictive biomarker for effective ICB response in NSCLC.

Previously, the group identified a unique tumor-infiltrating CD8+ T cell population with high PD-1 expression, dubbed PD-1T TILs (Thommen Nature Medicine 2018 [1]). These cells expressed markers of exhaustion but were highly clonal with tumor recognition capacity. Notably, their presence in pretreatment biopsies correlated with anti-PD-1 treatment response. In the current work, the team assessed whether this cell population possessed predictive capacity as a bona fide biomarker for ICB response.

To begin, Hummelink et al. collected baseline tumor specimens from two cohorts of late-stage NSCLC patients who later were treated with anti-PD-1 ICB (nivolumab or pembrolizumab). Formalin-fixed, paraffin-embedded tissue samples were stained and imaged using a digital, automated platform for PD-1T TIL quantification. The researchers segmented patients into training (n = 43) and validation (n = 77) cohorts, and tested the relationship between pre-treatment PD-1T TIL density (cells/mm2 in the section) and treatment response (disease control after 6 months). In the training group, PD-1T TIL density, specifically above or below 90 PD-1T TILs/mm2, predicted 6-month disease control with 79% sensitivity and 83% specificity. These findings were replicated in the validation cohort, in which PD-1T TIL levels also effectively predicted patient outcomes. Here, the PD-1T TIL density was >3-fold greater in ICB-responsive patients, and sensitivity and specificity of this biomarker were again high (77% and 67%). Importantly, across cohorts the team found that low PD-1T TIL density in pretreatment tumor specimens successfully indicated ICB non-responders.

Next, the researchers considered whether PD-1T TIL density could serve as a biomarker for additional treatment outcomes, such as 12-month disease control and survival metrics. Excitingly, disease control at 12 months was also predicted by PD-1T TIL levels (AUC = 0.78-0.89 and 92-93% sensitivity). In fact, the vast majority (97+%) of patients controlling disease at 12 months stratified in the high density (>90 PD-1T TILs/mm2) group. Progression-free and overall survival were also significantly higher in patients with high baseline PD-1T TIL density, compared to PD-1T-low patients.

ICB can lead to variable responses across tumor lesions in the same patient, and patients who experience progression of any lesion, even if others regress, are included in the progressive disease (PD) label based on RECIST criteria. Hence, the team next evaluated lesion-specific progression and PD-1T TIL density. This lesion-level characterization provided insight into local responses. Interestingly, among a set of 25 PD (12 month) patients with twice-measured CT responses, PD-1T-high biopsied lesions largely did not progress (~27%) compared to PD-1T low lesions (71%) in PD patients. Thus, local levels of PD-1T TILs may be predictive of ICB response on an individual lesion basis.

The authors next tested whether variables associated with tumor pathology or sample collection could influence the predictive ability of PD-1T TIL density. Promisingly, PD-1T TIL density was relatively consistent when sampling from different sites across a tumor, suggesting that even a small sample area could inform a tumor’s overall PD-1T TIL level. Further, clinicopathological features (such as adeno/squamous pathology, KRAS mutation status) or sample site (primary or metastatic) did not significantly alter PD-1T TIL density. However, the predictive value of PD-1T TILs was enhanced when samples were collected immediately prior to initiation of ICB, as opposed to before a previous line of therapy, and in tumor resection samples, versus biopsies.

Because PD-L1 levels have previously been used as a biomarker for ICB response, Hummelink et al. next compared the predictive capacities of PD-L1 expression and PD-1T TIL density. The PD-1T TIL metric (>90 PD-1T TILs/mm2) better predicted 6 and 12-month disease control compared to PD-L1 levels (either ≥50% or ≥1% expression). Additionally, adding PD-L1 expression analysis to PD-1T TIL density as dual biomarkers did not improve predictive ability over PD-1T TIL density alone.

Finally, the researchers assessed whether the presence of tertiary lymphoid structures (TLS) could indicate treatment outcomes. Previously, the team found that PD-1TILs were associated spatially with TLS, and in fact secreted CXCL13, a B cell chemoattractant involved in TLS formation. Based on CD3 and CD20 co-staining density, the authors identified TLS (> 60,000 dul cells per µm2) in ~1/3 of tumors, and TLS along with lower density lymphoid aggregates (LA; 10,000 - 60,000 cells per µm2) in about 1/2. As expected, PD-1T TIL density was greater within TLS relative to non-TLS tumor tissue, and overall PD-1T TIL numbers were reduced in tumor sections lacking TLS, highlighting their association. However, TLS were found relatively consistently between PD-1T-high and low tumors, and PD-1T-high tumors had more PD-1T TILs both inside and outside TLS. Overall, PD-1T TIL density was more effective than TLS presence as a biomarker for ICB response.

Taken together, the researchers found that patients with low baseline PD-1T TIL density largely do not respond to anti-PD-1 therapy, highlighting a population for which ICB may not be an effective treatment. Identifying key populations of responders or non-responders may reduce overtreatment, avoiding substantial cost and serious side effects. Moving forward, further investigation of PD-1T TILs in different tumor types and mechanistic study of their role in the ICB response may support immunotherapy discovery and tailored application for more effective cancer treatment.

[1] Thommen, D.S., Koelzer, V.H., Herzig, P. et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med 24, 994–1004 (2018). https://doi.org/10.1038/s41591...

Write-up by Alex Najibi, image by Lauren Hitchings

Meet the researcher

This week, first author Karlijn Hummelink and lead author Daniela Thommen answered our questions.

On the left, Karlijn Hummelink and on the right Daniela Thommen


What was the most surprising finding of this study for you?
As PD-1 blockade therapy is only effective in approximately 30% of advanced lung cancer patients, predictive biomarkers are urgently needed to improve clinical treatment decision making. Therefore, we wanted to identify a biomarker that allows us to better select the patients who respond to this therapy. Surprisingly, we found that, whereas PD-1T TILs could distinguish patients with long-term benefit to PD-1 blockade, they were even better in very accurately identifying a patient group without any benefit. Therefore, this biomarker should allow for reduce overtreatment and toxicities while minimizing undertreatment.

What is the outlook?

Our retrospective study established PD-1T TILs as a novel predictive biomarker for durable clinical benefit to PD-1 blockade in lung cancer patients. Next steps would include validating this biomarker in larger prospective trials as well as in other cancer types. Furthermore, we would like to explore methods that could improve standardization across centers and can be routinely used in any hospital. For example, we are currently working on the development of a gene expression signature that measures the presence of PD-1T TILs in a tumor and that could be easily screened for each patient in a diagnostic biopsy.

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

KH:
Recently I visited Norway for the first time and experienced the beautiful fjords, waterfalls, and hiking trails. For me it was the perfect place to relax and clear my mind after my first year as a pathology resident. I would definitely recommend to go hiking and camping in this impressive landscape to fully recharge your battery.
DT:
I recently visited the historical wine cellar of the Würzburg Residence and learned about their legendary wine from 1540. This wine resulted from an unusually hot year and was considered the best wine in a millennium. It is also the oldest existing wine; the second to last bottle was opened in 1961 after 421 years and was still drinkable! (at least the first two sips…). Now only one last bottle of this legendary wine is left.

References:

Hummelink K, van der Noort V, Muller M, Schouten RD, Lalezari F, Peters D, Theelen WSME, Koelzer VH, Mertz KD, Zippelius A, van den Heuvel MM, Broeks A, Haanen JBAG, Schumacher TN, Meijer GA, Smit EF, Monkhorst K, Thommen DS. PD-1T TILs as a predictive biomarker for clinical benefit to PD-1 blockade in patients with advanced NSCLC. Clin Cancer Res. 2022 Jul 19.

In the Spotlight...

Lung fibroblasts facilitate pre-metastatic niche formation by remodeling the local immune microenvironment

Gong and Li et al. described a COX-2+ lung adventitial fibroblast population, constitutively expressing high levels of inflammatory genes, that induced myeloid cells to be dysfunctional or immunosuppressive, fostering a pre-metastatic lung niche at steady state and enhanced by a distal tumor. In murine breast cancer models, COX-2 gene deletion in fibroblasts reversed myeloid cell dysfunction induced by IL-1β, PGE2, and other tumor-associated inflammation; boosted immune activation; and decreased lung metastasis. Genetic or pharmacologic inhibition of the PGE2 signaling pathway enhanced anti-metastatic activities of DC-based vaccines or PD-1 blockade.

Contributed by Paula Hochman

Targeted inducible delivery of immunoactivating cytokines reprograms glioblastoma microenvironment and inhibits growth in mouse models

Birocchi et al. engineered hematopoietic stem cells for targeted and inducible release of  IFNɑ or IL-12 by their monocyte/macrophage progeny at the tumor site and evaluated their efficacy, tolerability, and specificity. Compared to systemic treatment, inducible IFNɑ/IL-12 expression within the TME spared most of the brain and periphery from cytokine toxicity, but achieved a robust and durable response, up to eradication, in a GBM mouse model. Targeted IFNɑ gene therapy inhibited angiogenesis, decreased T cell exhaustion, and reprogramed TAMs toward a proinflammatory state while suppressing a mouse and human protumoral signature.

Contributed by Shishir Pant

Colocalized targeting of TGF-β and PD-L1 by bintrafusp alfa elicits distinct antitumor responses

Lan, Yeung, and Huang et al. investigated targeting TGFβ locally to PD-L1+ cells in the TME. Bintrafusp alpha (BA), a bifunctional TGFβRII–anti-PD-L1 fusion protein, was compared with fresolimumab (anti-TGFβ) or a combination of TGF-βRII (TGF-β trap) and anti-PD-L1 in multiple assays, including scRNAseq of MC38 tumors. BA exhibited an ~80-fold greater affinity due to avidity effects and PD-L1-dependent degradation of TGFβ following internalization. In MC38 tumors, BA increased CD4+ and CD8+ TILs, which correlated with superior efficacy and enhanced immune-related gene signatures (M1 MΦ), while decreasing TGFβ-driven pathways (EMT, fibrosis).

Contributed by Katherine Turner

Multiepitope supramolecular peptide nanofibers eliciting coordinated humoral and cellular antitumor immune responses

Wu et al. generated supramolecular α-helical peptide nanofiber vaccines including an EGFRvIII receptor B cell peptide epitope, a melanoma-associated TRP CD8+ T cell peptide epitope, and a tetanus toxoid CD4+ T cell epitope. Relative to CFA-peptide emulsions, the nanofiber multi-targeting vaccine induced comparable T cell responses and higher titers of specific antibodies, which mediated tumor lysis and phagocytosis by cells in vitro. In s.c. EGFRvIII+ melanoma mouse models, prophylactic and therapeutic vaccination induced antitumor effects exceeding those induced by controls; these effects were improved by anti-PD-L1 + anti-CD47 blocking antibodies.

Contributed by Paula Hochman

Intratumoral oncolytic herpes virus G47∆ for residual or recurrent glioblastoma: a phase 2 trial

Todo et al. conducted a single-arm, phase II trial of a mutated oncolytic herpes virus (G47Δ) administered intratumorally in glioblastoma that recurred or was incompletely resected. Adverse events were primarily immune-related, likely in response to the viral load. Although radiographic responses were limited, the overall 1-year survival in 13 patients at 12 months was 92%, prompting early termination of the trial and regulatory approval of the product in Japan. The 1-year survival was 84% in the 19 patients in the full analysis set; median OS was nearly 29 months. Neither IDH1 nor MGMT status affected outcomes (post-hoc), and TILs increased with dosing and persisted.

Contributed by Ed Fritsch

Personalized tumor vaccine for pancreatic cancer

Orr et al. examined the capacity of DC/tumor cell fusions to induce immune responses against pancreatic cancer cells. Compared to unvaccinated controls, mice vaccinated with a fusion of KPC cells (isolated from spontaneous pancreatic tumors) and bone marrow-derived DCs had slower tumor growth, improved survival time, and increased tumor recognition by T cells upon post-euthanasia ex vivo stimulation. In the PAN02 pancreatic cancer model, tumor/DC fusions elicited specific tumor immunity in vitro and in vivo. Patient-derived autologous DC/tumor fusions induced tumor-specific CD4+ and CD8+ activation upon ex vivo stimulation.

Contributed by Margot O’Toole

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.