Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Sep 1.
Published in final edited form as: J Immunother. 2021 Sep 1;44(7):248–253. doi: 10.1097/CJI.0000000000000372

Pretreatment Eosinophil Counts in Patients with Advanced or Metastatic Urothelial Carcinoma Treated with Anti–PD-1/PD-L1 Checkpoint Inhibitors

Jose Mauricio Mota 1,*,#, Min Yuen Teo 1,3,*, Karissa Whiting 2, Han A Li 3, Ashley Marie Regazzi 4, Chung-Han Lee 1,3, Samuel A Funt 1,3, Dean Bajorin 1,3, Irina Ostrovnaya 2, Gopa Iyer 1,3, Jonathan E Rosenberg 1,3
PMCID: PMC8373810  NIHMSID: NIHMS1700609  PMID: 34081050

Abstract

Eosinophils influence antitumor immunity and may predict response to treatment with immune checkpoint inhibitors (ICIs). To examine the association between blood eosinophil counts and outcomes in patients with advanced or metastatic urothelial carcinoma (mUC) treated with ICIs, we identified two ICI-treated cohorts: discovery (n=60) and validation (n=111). Chemotherapy cohorts were used as comparators (first-line platinum-based chemotherapy, n=75; second-line or more pemetrexed, n=77). The primary endpoint was overall survival (OS). Secondary endpoints were time on treatment (ToT) and progression-free survival. Univariate and multivariate analyses were performed using Cox proportional hazard models. Associations between changes in eosinophil count at weeks 2/3 and 6 after the start of ICI treatment were analyzed using landmark analyses. Baseline characteristics of the ICI cohorts were similar. In the discovery cohort, an optimal cutoff for pretreatment eosinophil count was determined (Eos-Lo: <100 cells/μL; n=9 [15%]; Eos-Hi: ≥100 cells/μL; n=51 [85%]). Eos-Lo was associated with inferior outcomes (OS: HR, 3.98 [95% CI, 1.85–8.56]; P<0.013; ToT: HR, 2.45 [95% CI, 1.17–5.10]; P=0.017). This was confirmed in the validation cohort (Eos-Lo: n=17 [15%]; Eos-Hi: n=94 [85%]) (OS: HR, 2.51 [95% CI, 1.31–4.80]; P=0.006; ToT: HR, 2.22 [95% CI, 1.29–3.80]; P=0.004), and remained significant after adjustment for other prognostic factors. Changes in eosinophil counts at weeks 2/3 and 6 were not clearly associated with outcomes. In chemotherapy cohorts, eosinophil counts were not associated with outcomes. In conclusion, low pretreatment eosinophil count was associated with poorer outcomes in patients with mUC treated with ICIs, and may represent a new predictive biomarker.

Keywords: metastatic urothelial carcinoma, bladder cancer, immunotherapy, eosinophils, predictive factor

INTRODUCTION

Anti–PD-1/PD-L1 immune checkpoint inhibitors (ICIs) are now established treatment options for metastatic urothelial carcinoma (mUC).15 Responses are potentially durable, although only 15% to 25% of patients with mUC exhibit response to ICIs.15 Various predictive and prognostic biomarkers are at different stages of evaluation, including disease-related factors (genomic alterations in DNA damage-repair [DDR] genes,6 tumor mutation burden, molecular subtype,7 and PD-L1 expression) and host-related factors (T-cell receptor clonality).8. Emerging data from other disease types indicate that non-T-cell–related immune cells, including eosinophils, might also be associated with ICI response.911

Eosinophils are granulocytes that are commonly elevated with parasitic infection, allergies, and autoimmune conditions.1213. Eosinophils were recently shown to be associated with the efficacy of ICIs in melanoma,10 lung cancer,9 and renal cell carcinoma.11 Peripheral blood eosinophil counts are part of the standard complete blood count panel and are therefore readily measurable and inexpensive to ascertain. We hypothesized that peripheral blood eosinophil count is associated with outcomes in patients with mUC treated with anti–PD-1/PD-L1 ICIs.

METHODS

Study Design and Patients

Our primary objective was to evaluate the association between peripheral blood eosinophil count and OS in patients with mUC treated with ICIs. Secondary objectives were to evaluate the association between peripheral blood eosinophil count and time on treatment (ToT) and progression-free survival (PFS), when data were available, and to explore the association between on-treatment changes in eosinophil counts and OS and ToT. The institutional review board at Memorial Sloan Kettering Cancer Center (MSK) approved this study before any procedures were conducted.

Four different cohorts of patients with mUC were included in this study. Two ICI-treated cohorts were included: a discovery cohort, consisting of patients enrolled in completed ICI clinical trials and reported previously in a study on DDR gene alterations,6 and a validation cohort, consisting of consecutive patients with mUC treated with ICIs as standard of care or in a clinical trial at MSK. In addition, two cohorts of patients treated with chemotherapy, which have previously been reported upon, were included: chemo cohort 1, consisting of patients with mUC treated with first-line platinum-based chemotherapy,14 and chemo cohort 2, consisting of patients with mUC treated with second-line or more pemetrexed.15 By design, there was no overlap between the discovery and validation cohorts.

Data Collection

Clinical data, including baseline and on-treatment complete blood count (CBC) results, were extracted from individual chart review. Pretreatment or baseline eosinophil counts were obtained from CBCs drawn within 7 days of the start of treatment, inclusive of the date of treatment. For on-treatment changes, we extracted eosinophil counts at the start of cycle 2 (i.e., at week 2 or 3 [±3 days], hereafter “week 2/3,” depending on the ICI agent used) and at week 6 (±3 days). Other clinical variables with known or proposed prognostic value were extracted, including variables comprising the Bellmunt prognostic classification, tumor mutational burden, and presence of mutations in DDR genes, as previously defined.6 All laboratory tests were performed in an MSK clinical laboratory.

Endpoints

The primary endpoint of this study was OS, defined as the time from the start of treatment to death or last follow-up. Secondary endpoints included ToT, which was evaluated for all cohorts, and PFS, which was evaluated for the discovery cohort only. ToT was defined as the interval between the first and last dose of ICI or chemotherapy. Patients still on treatment at the time of data cutoff were censored at the date of the last follow-up. PFS was defined as the time from the start of treatment to radiological progression (RECIST 1.1), death, or last follow-up. Data cutoff was January 15th, 2019.

Statistical Methods

Descriptive statistics were used to summarize patient characteristics in each cohort. To evaluate associations between eosinophil counts and outcomes of interest, eosinophil counts were analyzed as a continuous variable; to account for potential nonlinear effects, eosinophil counts were grouped and analyzed as a categorical variable. For categorization of eosinophil counts into high and low groups, we determined an optimal cutoff point using the discovery cohort by identifying the cut-off using the maxstat Lau94 package for R which provided the largest difference in OS. Time-to-event variables were estimated and plotted using the Kaplan-Meier method. Univariate Cox proportional hazard models were fit to evaluate associations between pretreatment eosinophil counts and OS, ToT, and PFS, and multivariate Cox models were used to adjust for other known prognostic factors. Rank-based correlation using an inverse probability censoring weighted estimator was performed to analyze Kendall’s tau between OS and PFS or ToT.

To assess associations between changes in eosinophil counts at week 2/3 and 6 and OS and ToT, landmark analyses at week 2/3 and 6 were conducted for the ICI cohorts. For these analyses, only subjects who had not experienced the event of interest before the week-2/3 or week-6 time point were considered, and that time point was treated as the new baseline time for survival analyses. We categorized the change in eosinophil count in two ways: (1) using an optimal cutoff point determined by maximizing the log-rank statistic, using OS as the endpoint in the initial cohort; and (2) by categorizing the change in eosinophil counts into three groups: decrease (change <0), no change (change=0), and increase (change >0). All analyses were conducted using R (version 3.6.1).

RESULTS

ICI Cohorts

The discovery cohort included 60 patients who started treatment between April 2014 and December 2016, and the validation cohort included 111 patients who started treatment between November 2014 and July 2018. Overall, the median age was 68 years (interquartile range, 60–76 years), 78% were male, and 71% had bladder as the primary tumor location. In total, 70% of patients received prior chemotherapy for metastases. The discovery cohort had a lower proportion of women (12% vs 27%; P=0.033) and higher baseline neutrophil counts (median, 5.6 vs 4.7 cells/μL; P=0.009). In the validation cohort, 19% of patients were treated with combined anti–PD-1 and anti-CTLA4 ICIs. Other baseline parameters, such as age, prior use of platinum chemotherapy, DDR status, tumor mutational burden, and Bellmunt prognostic group were comparable between the two cohorts (Table 1). OS was similar between the two cohorts (log-rank P=0.58), but ToT was significantly shorter in the validation cohort than in the discovery cohort (log-rank P<0.001) (Supplementary Figure 1).

Table 1.

Baseline characteristics of the immunotherapy cohorts

Characteristic Discovery Cohort
(n=60)
Validation Cohort (n=111) P
Age at treatment start, years 67 (61–72) 68 (60–76) 0.440
Sex 0.033
 Male 53 (88) 81 (73)
 Female 7 (12) 30 (27)
Primary site at diagnosis 0.800
 Bladder 45 (75) 77 (69)
 Renal pelvis/ureter 13 (22) 29 (26)
 Urethra 2 (3) 5 (5)
Metastatic sites
 Bone 19 (32) 28 (25) 0.500
 Liver 16 (27) 23 (21) 0.400
 Lung 20 (33) 47 (42) 0.300
 Nodes 41 (68) 79 (71) 0.700
Previous chemotherapy treatment for advanced disease >0.900
 Yes 42 (70) 78 (70)
 No 18 (30) 33 (30)
Hematologic parameters, cells/mL
 Eosinophil 0.10 (0.10–0.20) 0.10 (0.10–0.20) 0.800
 Lymphocyte 1.30 (0.88–1.70) 1.20 (0.85–1.60) 0.400
 Neutrophil 5.60 (4.40–7.45) 4.70 (2.60–6.20) 0.009
 Neutrophil:lymphocyte ratio 4.3 (3.30–6.30) 3.8 (2.60–6.20) 0.200
DDR Status
 Wild type 32 (53) 57 (51) 0.800
 Deleterious 15 (25) 25 (23)
 VUS 13 (22) 29 (26)
Tumor mutation burden, Mutations/Mb 7.3 (0.0 – 200.9) 7.8 (0.8 – 56.3) 0.233
Current treatment <0.001
 Anti–PD-1 43 (72) 55 (50)
 Anti–PD-L1 17 (28) 35 (32)
 Anti-CTLA4 + anti–PD-1 0 (0) 21 (19)
Bellmunt prognostic factors a 0.386
 0 10 (17) 28 (25)
 1 31 (52) 55 (50)
 2 or 3 19 (32) 28 (25)
a

Presence of liver metastasis, hemoglobin level of less than 10 g/dL, and ECOG performance status of more than zero.

Abbreviations: Data are median (interquartile range) or no. (%). DDR: DNA Damage response and repair genes; VUS: variant of unknown significance

Pretreatment Eosinophil Counts and Outcomes

In the discovery cohort, no significant associations between pretreatment eosinophil counts and other known variables were observed, including age, sex, Eastern Cooperative Oncology Group performance status, presence of hepatic or other visceral metastasis, Bellmunt prognostic group, prior treatment with platinum-based chemotherapy, type of immunotherapy received, DDR status, or tumor mutational burden.

Pretreatment eosinophil count as a continuous variable was positively associated with OS (HR, 0.01 [95% CI, 0.00–0.29]; P=0.009). The optimal cutoff for eosinophil count at baseline was determined to be 100 cells/μL, and patients were divided into two groups: Eos-Lo (<100 cells/μL; n=9 [15%]) and Eos-Hi (≥100 cells/μL; n=51 [85%]). Eos-Lo patients had significantly shorter OS than Eos-Hi patients (median OS, 3.5 vs. 22.4 months; log-rank P adjusted for multiple comparisons in cutoff-point selection using maxstat Lau94 approximation = 0.013) (Figure 1). On univariate analysis, DDR status, tumor mutational burden, and Bellmunt score (of 2 – 3) were also significantly associated with OS (Table 2). On multivariate analysis, eosinophil count remained significantly associated with OS after adjustment for DDR status and Bellmunt score (Table 3).

Figure 1. Outcomes in the discovery cohort.

Figure 1.

In the discovery cohort, patients with low baseline blood eosinophil counts (Eos-Lo; Eos <100 cells/μL) had shorter (A) time on ICI treatment and (B) overall survival than patients with high baseline blood eosinophil counts (Eos-Hi; ≥100 cells/μL).

Table 2.

Univariate analysis of overall survival in the immunotherapy cohorts

Characteristic Discovery Cohort Validation Cohort
HR 95% CI P HR 95% CI P
Age at treatment start, continuous variable 1.00 0.97–1.03 >0.900 0.99 0.97–1.02 0.500
Sex, male vs female (reference) 0.89 0.32–2.53 0.800 0.90 0.50–1.65 0.700
Prior chemotherapy treatment
 Prior platinum 1.05 0.53–2.10 0.900 1.81 0.97–3.38 0.062
 Any chemotherapy 1.12 0.55–2.26 0.700 1.94 1.00–3.77 0.049
Type of immunotherapy
 Anti–PD-1/anti–PD-L1 (reference) vs anti-CTLA4 ± anti–PD-1 NA NA NA 0.87 0.44–1.72 0.700
 Anti–PD-1 vs anti–PD-L1 (reference) 1.23 0.61–2.50 0.600 0.81 0.42–1.55 0.500
Pretreatment eosinophil counts
 Continuous variable 0.01 0.00–0.29 0.009 0.28 0.04–2.18 0.200
 Eos-Lo vs Eos-Hi (reference) 3.98 1.85–8.56 0.013 b 2.51 1.31–4.80 0.006
DDR status, DDRmut (deleterious + VUS) vs DDRwt 0.30 0.12–0.74 0.009 0.93 0.49–1.75 0.800
Tumor mutational burden 0.95 0.91–1.00 0.035 0.98 0.94–1.01 0.200
Bellmunt prognostic factors a
 1 vs 0 1.49 0.56–4.02 0.400 1.28 0.61–2.65 0.500
 2 or 3 vs 0 2.56 0.91–7.21 0.076 2.56 1.18–5.57 0.018
a

Presence of liver metastasis, hemoglobin level of less than 10 g/dL, and ECOG performance status of more than zero.

b

Log-rank P adjusted for multiple comparisons owing to cutoff point selection.

Abbreviations: DDR, DNA damage repair; DDRmut, any mutation in DDR; DDRwt, wild-type DDR; NA, not available; NLR, neutrophil-to-lymphocyte ratio; VUS, variants of unknown significance

Table 3.

Multivariate analysis of overall survival in immunotherapy cohorts

Characteristic Discovery Cohort Validation Cohort
HR 95% CI P HR 95% CI P
Pretreatment eosinophil count, Eos-Lo vs Eos-Hi (reference) 3.23 1.42–7.38 0.005 2.32 1.20–4.48 0.012
DDR status, DDRmut (deleterious + VUS) vs DDRwt 0.37 0.18–0.75 0.006 0.82 0.46–1.46 0.510
Bellmunt prognostic factors a
1 vs 0 1.10 0.40–3.02 0.860 1.25 0.59–2.64 0.567
2 or 3 vs 0 1.43 0.48–4.29 0.524 2.53 1.11–5.80 0.028
a

Presence of liver metastasis, hemoglobin level of less than 10 g/dL, and ECOG performance status of more than zero.

Abbreviations: DDR, DNA damage repair; DDRmut, any mutation in DDR; DDRwt, wild-type DDR

ToT was also shorter in the Eos-Lo group than in the Eos-Hi group (median, 2.3 vs 8.9 months; HR, 2.45 [95% CI, 1.17–5.10]; P=0.017) (Figure 1 and Supplementary Table 1). The association between baseline eosinophil count and ToT however lost its statistical significance after adjustment for DDR and Bellmunt risk status (Supplementary Table 2). Additionally, PFS was shorter in the Eos-Lo group than in the Eos-Hi group (HR, 3.11 [95% CI, 1.44–6.73]; P=0.004). OS was strongly correlated with both PFS and ToT (Kendall’s tau 0.78 and 0.76, respectively).

Analysis of the validation cohort revealed similar findings. Eos-Lo patients (n=17 [15%]) had shorter OS than Eos-Hi patients (n=94 [85%]) (median OS, 8.7 vs 21.6 months; HR, 2.51 [95% CI, 1.31–4.80]; P=0.006) (Figure 2). ToT was also shorter in Eos-Lo patients than in Eos-Hi patients (median, 2.4 vs 4.2 months; HR, 2.22 [95% CI, 1.29–3.80]; P=0.004) (Figure 2, Table 2, Supplementary Table 1, Supplementary Table 2). Association between eosinophil levels and ToT remained significant in this cohort after controlling for DDR and Bellmunt score.

Figure 2. Outcomes in the validation cohort.

Figure 2.

In the validation cohort, patients with low baseline blood eosinophil counts (Eos-Lo; Eos < 100 cells/μL ) had shorter (A) time on ICI treatment and (B) overall survival than patients with high baseline blood eosinophil counts (Eos-Hi; ≥100 cells/μL).

Early Changes in Eosinophil Counts

Further analyses were performed to investigate whether changes in peripheral blood eosinophil counts at week 2/3 and 6 after the start of ICI treatment were associated with differential OS and ToT. This analysis was performed using two approaches: (1) a landmark analysis at week 2/3 and 6, with the optimally selected cutoff point for each time point determined using the discovery cohort; and (2) a comparison of levels at week 2/3 and 6 with baseline levels, which was classified into three possible categories: decreased, unchanged, and increased. In the discovery cohort, at week 2/3, 3 patients were excluded (missing lab values, n=2; follow-up time <3 weeks, n=1). At 6 weeks, 11 patients were excluded because of missing values (evaluable, n=49). In the validation cohort, 10 and 24 patients were excluded from analyses at week 2/3 and 6, respectively, because of missing values.

There was a correlation between baseline eosinophil count and change at week 2/3 (Pearson correlation = −0.64; P<0.001) (Supplementary Table 3), indicating that any effect of change could be related to residual baseline effects. The optimally selected cutoff for change in eosinophil count was <100 versus ≥100 cells/μL (determined on the basis of OS in the discovery cohort). In the discovery cohort, at week 2/3, an increase in the eosinophil counts above the established cutoff was associated with shorter ToT (HR, 4.05; P=0.020), and this finding was confirmed in the validation cohort (HR, 1.78; P=0.040). Increased counts at week 2/3 were associated with shorter ToT than decreased counts (HR, 2.52 [95% CI, 1.01–6.30]; P=0.047), and unchanged counts were associated with shorter ToT than decreased counts, although this association was not statistically significant (HR, 2.03 [95% CI, 0.87–4.76]; P=0.100). A similar signal was observed in the validation cohort. Unchanged counts at week 2/3 were associated with shorter ToT than decreased counts (HR, 1.80 [95% CI, 1.05–3.08]; P=0.032), and increased counts were associated with shorter ToT than decreased counts, although this association was not statistically significant (HR, 1.50 [95% CI, 0.89–2.54]; P=0.13). Changes at week 2/3 were not associated with OS. No associations between changes at week 6 and ToT or OS were observed (Supplementary Table 3).

Chemotherapy Cohorts

To test whether the observations to date were mUC-related or therapy-specific features, we further evaluated the association between baseline eosinophil counts and outcomes in two separate chemotherapy-treated cohorts. Chemo cohort 1 included 75 patients with mUC treated with platinum-based chemotherapy as first-line treatment between October 2012 and September 2015.14 Chemo cohort 2 consisted of 77 patients treated with pemetrexed in the second-line (47%), third-line (48%), or fourth-line (16%) setting between November 2008 and November 2013.15 Baseline characteristics are presented in Supplementary Table 4. Of note, in chemo cohort 1, 7 of 71 evaluable patients (10%) were classified as Eos-Lo; in chemo cohort 2, 33 of 77 evaluable patients (43%) were classified as Eos-Lo.

For chemo cohort 1, no associations between pretreatment eosinophil counts and OS were observed, both when eosinophil count was considered as a continuous variable (HR, 0.53 [95% CI, 0.08–3.76]; P=0.500) and when separated as categorical values using previously determined cutoffs (HR, 0.82 [95% CI, 0.29–2.31]; P=0.700). Similarly, no associations between pretreatment eosinophil counts and ToT (categorical: HR, 0.55 [95% CI, 0.25–1.21]; P=0.140) or PFS (categorical: HR, 1.06 [95% CI, 0.45–2.49]; P=0.900) were observed (Supplementary Table 5, Supplementary Figure 2). Similar outcomes were observed for chemo cohort 2, in which pretreatment eosinophil counts were not associated with either OS (categorical: HR, 0.93 [95% CI, 0.57–1.50]; P=0.800) or PFS (categorical: HR, 0.74 [95% CI, 0.47–1.17]; P=0.200) (Supplementary Figure 3).

DISCUSSION

ICIs are an established treatment option for advanced urothelial carcinoma, and their efficacy may be influenced by a series of disease- and host-related factors.8 To our knowledge, this is the first work to show that a low pretreatment peripheral eosinophil count (observed in ~15% of patients with mUC) is independently associated with significantly shorter OS among patients with mUC treated with ICI. The correlation was demonstrated in two independent ICI-treated cohorts. To further demonstrate that this was an ICI-specific phenomenon, we further evaluated two separate chemotherapy cohorts—no association was observed between eosinophil count and clinical outcomes in these cohorts.

Eosinophils are specialized granulocytes that are part of the innate immune system and are involved in many biological processes, including allergic response, autoimmune conditions, and response to parasitic infections.12,16 Preclinical models have associated eosinophils with tumor immune surveillance, antitumor immunity, suppression of metastasis, and antitumor cytotoxicity,13,1721 but correlations with clinical outcomes have remained unclear.2224 Immunomodulatory therapies—including high-dose IL-2 25 and, more recently, anti-CTLA4 26 and anti–PD-1 blockade 10—have long been shown to induce eosinophilia. Various reports across different disease types, including melanoma,10, non-small cell lung cancer,9 and renal cell carcinoma,11 have found an association between higher peripheral eosinophil count and a lower risk of progression or longer OS. Our observations of patients with mUC were not dissimilar from prior observations of patients with different tumor types. In two ICI cohorts, low pretreatment eosinophil levels were consistently associated with shorter OS, independent of DDR status and Bellmunt score. Additionally important is that no association was observed in two separate cohorts of patients treated with cytotoxic chemotherapy, confirming that eosinophil count is in fact a predictive factor, instead of a general prognostic indicator.

On-treatment peripheral eosinophil kinetics did not provide additional predictive or prognostic value in our data set. This is in contrast to the protective benefit seen with early increases in eosinophil counts with ipilimumab in patients with advanced melanoma 26 and other treatment-related immune cell alterations.27,28 We did, however, observe an early increase in peripheral eosinophil counts at week 2/3, which might be associated with shorter ToT, although the observation was not consistent across cohorts and methods of analyses, nor was it correlated with OS. It is unclear whether this lack of association is attributable to the limited sample size or whether this is a biological factor.

In our analysis, we observed that while pretreatment eosinophil levels and DDR alterations were independent predictors of OS in the discovery cohort, DDR alterations were not significantly associated with outcomes in the validation cohort. This could be secondary to inherent biological and clinical differences between clinical trial and standard of care populations, as seen in different ToT between both cohorts (Supplementary Figure 1).

Although our analysis is limited by its retrospective nature, the inclusion of an ICI validation cohort and two chemotherapy cohorts added robustness to this analysis. Furthermore, eosinophil count is part of the standard complete blood count panel, and therefore levels were prospectively obtained as patients received their treatment.

Our observations suggest that baseline eosinophil count lower than 100 cells/μL is inversely associated with OS in patients receiving ICIs. We did not observe a correlation between elevated eosinophil counts and clinical outcomes. However, eosinophilia has been associated with high-grade immune-related cutaneous adverse events,29 and immune-related toxicity has, in turn, been associated with treatment efficacy,30 suggesting that eosinophils play a complex and poorly understood role in the efficacy and toxicity of ICIs. As a biomarker with potential clinical utility, baseline blood eosinophil count can be assessed easily and at low cost with an established clinical-grade assay. If our findings are prospectively validated, pretreatment blood eosinophil counts should be integrated into larger-scale nomograms or biomarker efforts to improve our ability to identify patients who are most likely to benefit, or not, from ICIs.

Supplementary Material

Supplementary data
Supplemental Data File_1
Supplemental Data File_2
Supplemental Data File_3

Acknowledgments:

David B. Sewell of the Memorial Sloan Kettering,

Department of Surgery provided editorial assistance.

REFERENCES

  • 1.Balar AV, Castellano D, O’Donnell PH, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol 2017;18(11):1483–92. [DOI] [PubMed] [Google Scholar]
  • 2.Balar AV, Galsky MD, Rosenberg JE, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet 2017;389(10064):67–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 2017;376(11):1015–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Powles T, Duran I, van der Heijden MS, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2018;391(10122):748–57. [DOI] [PubMed] [Google Scholar]
  • 5.Powles T, O’Donnell PH, Massard C, et al. Efficacy and safety of durvalumab in locally advanced or metastatic urothelial carcinoma: updated results from a phase 1/2 open-label study. JAMA Oncol 2017;3(9):e172411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Teo MY, Seier K, Ostrovnaya I, et al. Alterations in DNA damage response and repair genes as potential marker of clinical benefit from PD-1/PD-L1 blockade in advanced urothelial cancers. J Clin Oncol 2018;36(17):1685–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kim J, Kwiatkowski D, McConkey DJ, et al. The Cancer Genome Atlas expression subtypes stratify response to checkpoint inhibition in advanced urothelial cancer and identify a subset of patients with high survival probability. Eur Urol 2019;75(6):961–4. [DOI] [PubMed] [Google Scholar]
  • 8.van Dijk N, Funt SA, Blank CU, et al. The cancer immunogram as a framework for personalized immunotherapy in urothelial cancer. Eur Urol 2019;75(3):435–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tanizaki J, Haratani K, Hayashi H, et al. Peripheral blood biomarkers associated with clinical outcome in non-small cell lung cancer patients treated with nivolumab. J Thorac Oncol 2018;13(1):97–105. [DOI] [PubMed] [Google Scholar]
  • 10.Weide B, Martens A, Hassel JC, et al. Baseline biomarkers for outcome of melanoma patients treated with pembrolizumab. Clin Cancer Res 2016;22(22):5487–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zahoor H, Barata PC, Jia X, et al. Patterns, predictors and subsequent outcomes of disease progression in metastatic renal cell carcinoma patients treated with nivolumab. J Immunother Cancer 2018;6(1):107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Huang L, Appleton JA. Eosinophils in Helminth infection: defenders and dupes. Trends Parasitol 2016;32(10):798–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ikutani M, Yanagibashi T, Ogasawara M, et al. Identification of innate IL-5-producing cells and their role in lung eosinophil regulation and antitumor immunity. J Immunol 2012;188(2):703–13. [DOI] [PubMed] [Google Scholar]
  • 14.Teo MY, Bambury RM, Zabor EC, et al. DNA damage response and repair gene alterations are associated with improved survival in patients with platinum-treated advanced urothelial carcinoma. Clin Cancer Res 2017;23(14):3610–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bambury RM, Benjamin DJ, Chaim JL, et al. The safety and efficacy of single-agent pemetrexed in platinum-resistant advanced urothelial carcinoma: a large single-institution experience. Oncologist 2015;20(5):508–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Diny NL, Rose NR, Cihakova D. Eosinophils in autoimmune diseases. Front Immunol 2017;8:484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Carretero R, Sektioglu IM, Garbi N, et al. Eosinophils orchestrate cancer rejection by normalizing tumor vessels and enhancing infiltration of CD8(+) T cells. Nat Immunol 2015;16(6):609–17. [DOI] [PubMed] [Google Scholar]
  • 18.Reichman H, Itan M, Rozenberg P, et al. Activated eosinophils exert antitumorigenic activities in colorectal cancer. Cancer Immunol Res 2019;7(3):388–400. [DOI] [PubMed] [Google Scholar]
  • 19.Simson L, Ellyard JI, Dent LA, et al. Regulation of carcinogenesis by IL-5 and CCL11: a potential role for eosinophils in tumor immune surveillance. J Immunol 2007;178(7):4222–9. [DOI] [PubMed] [Google Scholar]
  • 20.Tepper RI, Coffman RL, Leder P. An eosinophil-dependent mechanism for the antitumor effect of interleukin-4. Science 1992;257(5069):548–51. [DOI] [PubMed] [Google Scholar]
  • 21.Tepper RI, Pattengale PK, Leder P. Murine interleukin-4 displays potent anti-tumor activity in vivo. Cell 1989;57(3):503–12. [DOI] [PubMed] [Google Scholar]
  • 22.Alrawi SJ, Tan D, Stoler DL, et al. Tissue eosinophilic infiltration: a useful marker for assessing stromal invasion, survival and locoregional recurrence in head and neck squamous neoplasia. Cancer J 2005;11(3):217–25. [DOI] [PubMed] [Google Scholar]
  • 23.Ownby HE, Roi LD, Isenberg RR, et al. Peripheral lymphocyte and eosinophil counts as indicators of prognosis in primary breast cancer. Cancer 1983;52(1):126–30. [DOI] [PubMed] [Google Scholar]
  • 24.Steel JL, Kim KH, Dew MA, et al. Cancer-related symptom clusters, eosinophils, and survival in hepatobiliary cancer: an exploratory study. J Pain Symptom Manage 2010;39(5):859–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Huland E, Huland H. Tumor-associated eosinophilia in interleukin-2-treated patients: evidence of toxic eosinophil degranulation on bladder cancer cells. J Cancer Res Clin Oncol 1992;118(6):463–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gebhardt C, Sevko A, Jiang H, et al. Myeloid cells and related chronic inflammatory factors as novel predictive markers in melanoma treatment with ipilimumab. Clin Cancer Res 2015;21(24):5453–9. [DOI] [PubMed] [Google Scholar]
  • 27.Snyder A, Nathanson T, Funt SA, et al. Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: an exploratory multi-omic analysis. PLoS Med 2017;14(5):e1002309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Vilain RE, Menzies AM, Wilmott JS, et al. Dynamic Changes in PD-L1 expression and immune infiltrates early during treatment predict response to PD-1 blockade in melanoma. Clin Cancer Res 2017;23(17):5024–33. [DOI] [PubMed] [Google Scholar]
  • 29.Phillips GS, Wu J, Hellmann MD, et al. Treatment outcomes of immune-related cutaneous adverse events. J Clin Oncol 2019;37(30):2746–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Maher VE, Fernandes LL, Weinstock C, et al. Analysis of the association between adverse events and outcome in patients receiving a programmed death protein 1 or programmed death ligand 1 antibody. J Clin Oncol 2019;37(30):2730–7. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary data
Supplemental Data File_1
Supplemental Data File_2
Supplemental Data File_3

RESOURCES