Page 40 - Read Online
P. 40

Briggs et al. J Cancer Metastasis Treat 2021;7:46  https://dx.doi.org/10.20517/2394-4722.2021.84  Page 9 of 13

                              low
                   high
               Teff vs. Myeloid , Teff GES in the JAVELIN Renal 101 cohort in either the avelumab + axitinib arm, or
                                     high
                                                                   high
               the sunitinib monotherapy arm, suggesting that the Myeloid , Teff subgroup may be most resistant to
                                                                          high
               ICI monotherapy rather than targeted monotherapy.
               While the prognostic and predictive value of these GES requires further validation, we found the strongest
               consensus for angiogenic GES (IMmotion 150 Angio, JAVELIN Renal 101 Angio) as biomarkers predictive
               of improved response to sunitinib and for immunogenic GES (IMmotion 150 Teff, JAVELIN Renal 101
               Immuno) as biomarkers predictive of improved response to ICI therapy. Additionally, myeloid
               inflammation GES (IMmotion Teff, Myeloid) may predict improved response to combination anti-VEGF +
               ICI therapy vs. ICI therapy alone. Table 4 and Supplementary Table 4 list all GES biomarkers associated
               with predictive or prognostic outcomes.


               PDL1 STATUS AS A PROGNOSTIC AND PREDICTIVE BIOMARKER
               As the principal biologic target of many of the ICIs, the expression of PDL1 on renal tumor cells has
               received significant attention as a potential prognostic and predictive biomarker. Prognostically, a meta-
               analysis in 2020 reported that PDL1 expression of tumor cells was positively associated with both OS (HR =
                                                                        [49]
               1.98, 95%CI: 1.22-3.22) and DFS (HR = 3.70, 95%CI: 2.07-6.62) . These findings are notable because
               tumors with high expression of PDL1 have been previously shown to demonstrate aggressive behavior [50-58] .
               The improved OS in PDL1-expressing tumors in the era of ICIs possibly occurs because PDL1 expression
               may also predict tumor response to immunotherapy.

               A recent 2020 meta-analysis included 4635 patients across six randomized controlled trials (RCTs)
               published before May 2018 with available PDL1 expression data and compared ICI vs. standard of care
               therapy (SOC). Regardless of PDL1 expression level, ICI therapy improved both PFS and OS compared to
               SOC. However, in PDL1 positive patients receiving ICI, PFS was improved vs. SOC (HR = 0.75, 95%CI:
               0.63-0.89, P < 0.0001) but OS was not (HR = 0.72, CI: 0.63-0.81, P = 0.63). Since this meta-analysis, two of
               the included RCTs have published longer-term follow-up data on the effect of PDL1 status on response to
               ICI without significant change to earlier-published data. Furthermore, other studies assessing response to
               ICI based on PDL1 status report both significant [Table 5] and non-significant [Supplementary Table 5]
               associations between differential PDL1 expression and PFS and OS.

               Overall, we found the strongest consensus for PDL1 as a prognostic biomarker for OS and PFS. Notably,
               PDL1 expression is dynamic. Therefore, the assessed tissues (primary tumor vs. metastasis) and timing of
               tissue acquisition (especially if primary tumor resection occurs long before evidence of metastasis) may
               impact PDL1 expression, and therefore the accuracy of assessment as a biomarker.  Table 5 and
               Supplementary Table 5 listed all identified data depicting the values of PDL1 as a prognostic or predictive
               biomarker associated with PFS, OS, or CSS.

               CONCLUSION
               We reviewed the serum, gene mutation, genetic expression, and histologic biomarkers that predict response
               to treatment and prognosticate clinical outcomes. Current survival models may be improved by
               incorporation of newly proven biomarkers, allowing providers to give more accurate and individualized
               prognosis to patients. Future predictive models may be built to allow oncologists to prescribe the most
               effective treatment regimens for an individual patient’s tumor and biologic profile. It is clear that patients
               with mRCC will benefit from continued measurement of biomarkers in large clinical trials assessing clinical
               responses to various treatment regimens in patients with mRCC, and their incorporation into increasingly
               personalized predictive tools.
   35   36   37   38   39   40   41   42   43   44   45