In this phase 2 study, we investigated the clinical outcomes and dynamic immune landscapes associated with regorafenib-nivolumab as first-line therapy in patients with unresectable HCC. The primary endpoint was met: the ORR was 31.0% with the median PFS of 7.38 months. These efficacy outcomes were comparable with the previously reported efficacy outcomes with other first-line anti-PD-1/PDL1-MKI combinations5–7 or atezolizumab-bevacizumab3. Overall, regorafenib-nivolumab was well-tolerated with previously known toxicity profiles, and no new safety signal with the combination. These findings were consistent with a previous phase 1 study of regorafenib-nivolumab in other gastrointestinal cancers18. The present results suggest that regorafenib-nivolumab may be a clinically feasible first-line treatment option in patients with unresectable HCC, warranting further investigation.
In the current study, the systematic collection of samples in a clinical trial setting provided a unique opportunity to explore the immune landscapes reprogrammed by regorafenib-nivolumab (summarized in Supplementary Table 3). To our knowledge, this is the first study to explore, at a single-cell level, the dynamic immune landscapes of HCC patients treated with first-line ICI-based combination therapy. We revealed the clinical relevance of diverging CD8+ T-cell and classical monocyte responses, which could provide novel specific evidence for future development of biomarkers and novel immunotherapeutic strategies in HCC patients.
Our finding that the characteristics of CD8+ T cells upon regorafenib-nivolumab treatment were associated with efficacy outcomes solidifies the importance of T-cell activation in the context of ICI-based treatment. The TCR repertoire diversification in long-term responders following regorafenib-nivolumab treatment suggests that favorable outcomes were associated with the clonal expansion of CD8+ T cells recognizing a variety of neoantigens to a level that was detectable in peripheral blood23–25. To our knowledge, this is the first such demonstration in HCC patients receiving ICI-based treatments. A proliferative burst of CD8+ T cells prominently seen among long-term responders accords well with previous studies dealing with its clinical value upon ICI-based treatment22,26. We also found that only long-term responders exhibited enrichment of genes representing ICI responsiveness and cytotoxicity in proliferating MKI67+ CD8+ T cells, which further directed classical monocytes through IFN-γ-related pathways. This previously under-recognized clinically relevant interaction observed in long-term responders suggests that these interactions could potentially be harnessed to further enhance anti-tumor response, and suggests that early progressors may have the presence of factors precluding effective T-cell responses, and subsequent monocyte activation, despite regorafenib-nivolumab treatment.
Diverging monocyte response according to clinical outcomes is the one of the unique aspects of the present study. It is increasingly recognized that myeloid lineage cells play a critical role in meticulously balancing anti-tumor responses and immuno-suppression in the context of ICI-based treatments. In HCC patients, we previously found that the increased frequency of classical monocytes upon nivolumab monotherapy was more prominent in patients with durable clinical benefit than in those without31. CD38+ macrophages have also been associated with favorable survival outcomes in HCC patients treated with anti-PD-1/PD-L132. Recent single-cell analysis demonstrated that the abundance of CD11c+ antigen-presenting cells was associated with the response to anti-PD-1 therapy in HCC patients33. In the present study, we showed interactions between proliferating MKI67+ CD8+ T cells and classical monocyte subsets, as well as a higher probability of M1-directed polarization in long-term responders. These results point to the clinical relevance of positive associations between IFN-γ response and myeloid populations. Of note, our results validate the clinical relevance of the IFN-γ-directed myeloid response in the context of anti-PD-1/PD-L1-MKI combinations, which warrants the development of relevant myeloid-related biomarkers in this setting.
Conversely, in early progressors, the signature of immunosuppressive tumor-associated macrophages was prominently enriched on M2-skewed non-classical monocytes overexpressing immune suppressive markers, such as CSF1R, SIGLEC10, and VSIR (encoding VISTA), despite regorafenib-nivolumab treatment. This accords well with a previous report that immunosuppressive Arg-1-expressing CD163+ macrophages were enriched in HCC patients not responding to cabozantinib-nivolumab34. To further examine these diverging monocyte responses in different clinical subgroups, we compared the gene expression profiles of monocyte subsets, which revealed that TMEM176A/B was associated with early disease progression. TMEM176A/B is a surface protein highly expressed on myeloid immune cells, which was recently shown to prevent inflammasome responses, thereby preventing effective ICI-mediated immune responses29. Accordingly, regorafenib-nivolumab did not sufficiently increase the expression levels of NLRP3-related genes on monocytic populations from early progressors, possibly because of upregulation of TMEM176A/B. This novel association between early disease progression and up-regulation of TMEM176A/B coupled with an ineffective inflammasome response suggests a possibility to develop novel immunotherapeutic strategies to augment the inflammasome-related response, to overcome primary resistance to ICI-based treatments in HCC patients.
The clinical benefits of ICI-based combinations suggest different additive and/or synergistic effects of the combination partners. In a study of the atezolizumab-bevacizumab combination, the addition of bevacizumab to atezolizumab led to decreases in VEGFR2 expression levels and increased regulatory T cells, leading to improved survival outcomes particularly in HCC patients with high expression of VEGFR2, increased frequency of regulatory T cells, and enrichement of myeloid inflammation signatures21. On the other hand, in a study of neoadjuvant cabozantinib-nivolumab in HCC patients, the addition of cabozantinib decreased the level of CXCL1, a chemokine ligand involved in immune-resistance, and subsequently promoted T-cell activation34. Cabozantinib also reportedly induces intratumoral neutrophil infiltration, which further enhances the inflammatory phenotype, when used in combination with nivolumab in preclinical models of HCC35. In addition to its anti-angiogenic activity, regorafenib can potentially modulate myeloid cell populations by inhibiting the CSF-1/CSF-1R pathway8,13, which drives the immunosuppressive gradient of myeloid cells. In our current study, we demonstrated regorafenib-specific effects on classical monocytes, specifically in long-term responders—which was characterized by enrichment of a regorafenib-induced gene signature and a gene signature representing CSF-1 deficient status, highlighting the clinical relevance of the immunomodulatory effect of regorafenib. The regorafenib-nivolumab combination is actively being explored in other cancer types (NCT04879368), and regorafenib plus anti-PD-1/L1 combinations are being investigated in HCC patients (NCT04718909, NCT04183088), supporting the clinical relevance and generalizability our present findings.
Alterations in the WNT/β-catenin pathway have been previously demonstrated to be associated with excluding anti-tumor immune responses, thereby conferring resistance to ICIs36,37. However, Our ctDNA analysis revealed that genetic alterations in the Wnt/β-catenin pathway was not associated with poor survival outcomes. Similarly, the survival outcomes were comparable between those with versus without a CTNNB1 (encoding β-catenin) mutation, among HCC patients treated with atezolizumab plus bevacizumab21. The available data indicate that there is no need to preclude patients with WNT/β-catenin pathway mutations from receiving ICI-based combinations involving anti-angiogenic agents. Mutations in the PI3K/mTOR pathway detected by ctDNA are reportedly associated with poor survival outcomes in HCC patients treated with TKIs, but with a trend towards favorable survival outcomes in patients treated with ICI38. In our analysis, patients with alterations in the PI3K/mTOR pathway showed a trend towards favorable OS compared to those without38. Given the small numbers of patients involved in these results, further studies are warranted to delineate the clinical implications of mutations in the context of ICI-based combinations.
The absence of a control group (i.e., nivolumab monotherapy) may be one of the limitations of the current study, precluding the accurate interpretation on the effects of adding regorafenib to nivolumab. Nevertheless, given that anti-PD-1/PD-L1 monotherapy was not a standard 1st line option at the time of study design, we did not include a control group involving nivolumab monotherapy from the study design. Moreover, the subsequently reported results of the CheckMate459 which failed to demonstrate the superiority of nivolumab over sorafenib 1, which also does not support the use of the nivolumab monotherapy control arm.
In conclusion, our present results demonstrate that regorafenib-nivolumab therapy has clinical activity and is well tolerated as a first-line treatment for patients with unresectable HCC. The differential quantitative and qualitative responses of CD8+ T cells and classical monocytes, and their interconnected relationships, upon regorafenib-nivolumab treatment highlight these cellular subsets as critical immune regulators in this therapeutic context. Modulation of the inflammasome response to overcome resistance to ICI-based treatments may be a potential novel immunotherapeutic approach that warrants further investigations in HCC patients.