Abstract
Introduction
Nivolumab, a programmed death 1 (PD-1) inhibitor, has recently demonstrated efficacy as second-line therapy for esophageal squamous cell carcinoma (ESCC) patients in a phase III trial. We report real-world clinical outcomes of nivolumab therapy for ESCC patients.
Methods
ESCC patients refractory/intolerant to at least one line of chemotherapy and who received nivolumab as a subsequent line of therapy were included. The efficacy and safety of nivolumab and the predictive role of PD-L1 and CD8 expression were analyzed.
Results
Fifty-eight patients were analyzed for safety and survival outcomes, while 57 were analyzed for objective response rates (ORR) excluding one with no measurable lesions. Eleven patients achieved a partial response, leading to an ORR of 19.3%. The median response duration was 6.5 months (range 4.1–22.4). The median progression-free survival (PFS) and overall survival were 2.1 (95% confidence interval [CI] 1.8–2.3) and 7.4 (95% CI 4.8–10.0) months, respectively. Among patients with adequate samples, 56.9% (29/51), 27.5% (14/51), and 17.6% (9/51) expressed a combined positive score (CPS) ≥ 1, ≥ 10, and ≥ 20, respectively, while 24.4% (11/45) and 57.5% (23/40) were positive for intratumoral and peritumoral CD8 + T cell infiltration, respectively. A significantly longer PFS was observed in patients with a CPS ≥ 20 (7.5 [95% CI 1.8–13.1] vs. 1.9 [1.4–2.3] months, P = 0.05), and a trend towards better survival was seen in those with CPS ≥ 10 or intratumoral CD8 + T cell infiltration.
Conclusions
Nivolumab is a valuable option at subsequent treatment lines for patients with advanced ESCC.
Electronic supplementary material
The online version of this article (10.1007/s00262-020-02766-7) contains supplementary material, which is available to authorized users.
Keywords: Esophageal squamous cell carcinoma, Nivolumab, PD-1 inhibitor, PD-L1 expression, CD8 expression
Introduction
Esophageal cancer ranked seventh in incidence (572,000 new cases) and sixth in mortality (509,000 deaths) worldwide in both sexes in 2018 [1]. Esophageal cancer consists mostly of two main histologic types: esophageal adenocarcinomas (ECAs) and squamous cell carcinomas (ESCCs).
While ESCC has been decreasing in incidence in North America and West Europe, it remains the highly prevalent histologic type of esophageal cancer in East Asia and Africa [2]. Among 2483 patients with newly diagnosed esophageal cancer in South Korea in 2017, 2255 (90.8%) cases were ESCC and 60 (2.4%) were ECAs [3]. This epidemiologic characteristic was similar to that from other East Asian countries where ESCC comprised over 90% of total cases [2, 4].
Cisplatin in combination with fluorouracil in first-line therapy, followed by taxane or irinotecan in second-line therapy, constitutes the standard treatment for advanced esophageal cancer. However, sequential chemotherapy results in disappointing outcomes with a median overall survival of less than 1 year [5].
Immune checkpoint inhibitors have revolutionized the treatment schemes across many cancer types since their first approval for melanoma in 2014 [6]. Anti-programmed cell death protein 1 (PD-1) antibodies inhibit the interaction between the PD-1 expressed on T cells and its ligands, programmed cell death ligand-1 (PD-L1) and programmed cell death ligand-2, expressed on antigen-presenting cells and tumor cells, thereby reverting the ability of T cells to recognize and kill tumor cells [7].
As expected, efforts to evaluate the efficacy of immunotherapy have expanded to include esophageal cancer. Pembrolizumab, a PD-1 inhibitor, proved its efficacy as second-line therapy for esophageal cancer in KEYNOTE-181 [8], and has been approved in the USA since August 2019 for patients with high PD-L1 expressing esophageal cancer.
Nivolumab, another well-known PD-1 inhibitor, was studied in a single-arm phase II trial (ATTRACTION-1) of 65 Japanese patients with ESCC [9]. The drug showed a promising efficacy with acceptable toxicity profiles in ESCC patients who had progressed after chemotherapy. Another landmark study (ATTRACTION-3) compared the efficacy of second-line nivolumab to that of conventional chemotherapy for ESCC patients [10], and successfully demonstrated an overall survival benefit in those treated with nivolumab. Based on these results, since February 2020, nivolumab has been approved in Japan for patients with ESCC that had progressed following chemotherapy.
In Korea, nivolumab was not endorsed for the treatment of esophageal cancer until official approval was granted in April 2020. However, based on the outcome of the ATTRACTION-1 trial, our institute received preliminary approval in November 2017 to use nivolumab in the treatment of patients whose cancer had progressed after fluoropyrimidine-, platinum-, or taxane-based chemotherapy. Here, we present the outcomes of nivolumab, with comprehensive analysis on the predictive role of PD-L1 and CD8 expressions, as second or further-line therapy for ESCC patients.
Materials and methods
Patients and data collection
We conducted a retrospective analysis on patients diagnosed with ESCC who were treated with nivolumab in second- or further-line therapy from November 2017 to November 2019. All patients were pathologically confirmed to have ESCC and refractory or intolerant to at least one line of fluoropyrimidine-, platinum- or taxane-based chemotherapy.
Nivolumab therapy
Patients received 3 mg/kg nivolumab intravenously every 2 weeks until disease progression, unacceptable toxic effects, or death. The Response Evaluation Criteria in Solid Tumors version 1.1 were used to assess the treatment response [11]. Adverse events (AEs) were assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE), version 4.0.
Immunohistochemistry (IHC)
PD-L1 expression was assessed in pre-treatment samples obtained from endoscopic biopsies or surgical resection. Assessment of PD-L1 expression was done by experienced pathologists at our institution. All samples except one, which was tested with PD-L1 22C3 pharmDx, were tested using a PD-L1 IHC SP263 kit on the DAKO ASL48 platform (Tucson, AZ) according to the manufacturer’s recommendations. The combined positive score (CPS) was calculated by summing the number of PD-L1-stained cells (tumor cells, lymphocytes, and macrophages) and dividing the result by the total number of viable tumor cells, multiplied by 100. The tumor proportion score (TPS) was calculated by summing the number of PD-L1-stained tumor cells and dividing the result by the total number of viable tumor cells, multiplied by 100 (supplementary Fig. 1) [12].
For the CD8 IHC test, paraffin-embedded tumor sections were dewaxed in xylene and ethanol, and autoclaved for 60 min in an antigen retrieval solution (CC1), and were incubated for 24 min in Ventana BenchMark XT with anti-CD8 (RTU; clone SP57; Ventana Medical Systems, Roche, Tucson, Arizona, USA). The secondary antibody was incubated with an ultraview universal DAB detection kit (Ventana Medical Systems, Roche, Tucson, Arizona, USA). A PD-L1 IHC test using a PD-L1 IHC 22C3 pharmDx kit on the DAKO ASL48 platform was performed according to the manufacturer’s recommendations.
CD8 + T cells were quantified by screening the representative cancer area using a modification of the previous methods as follows [13]. For the quantitative evaluation of intratumoral and peritumoral CD8 + T cells in representative sections, we chose three microscopic fields, counted intratumoral/peritumoral CD8 + T cells at × 200 magnification, and averaged them respectively (supplementary Fig. 2). CD8 + T cell infiltration was considered positive when the average number of CD8 + T cells was > 50 [13].
Statistical analysis
All patients were included for survival analysis, and only those with measurable lesions were included for response analysis. The comparison of objective response rates (ORR) according to specific PD-L1 expression levels was performed using the Fisher’s exact test or χ2 test. Progression-free survival (PFS) was defined as the time from initiation of nivolumab to the documentation of either disease progression or death. Overall survival was defined as the time from the initiation of nivolumab to death from any cause. Survival curves were calculated using the Kaplan–Meier method and compared using the log-rank test. Patients were censored at the time of analysis if they were progression-free and/or alive. All analyses were performed using IBM SPSS Statistics 25 (Armonk, NY), and a P value of less than 0.05 was considered statistically significant. The data cutoff date was January 31, 2020.
Results
Patient characteristics
A total of 58 patients diagnosed with ESCC who had received at least one cycle of nivolumab as second- or further-line therapy were included in this study. Table 1 shows patient characteristics. The median age was 65 (range 39–85) and most patients were male and had good Eastern Cooperative Oncology Group (ECOG) performance status (PS), while five patients had ECOG PS 2 or 3. Fifty percent of patients received one prior chemotherapy, whereas 37.9% received two, and 12.1% received three or more. Additionally, 44.8% and 31.0% underwent surgery and concurrent chemoradiation, respectively, with curative intent.
Table 1.
Characteristics | N = 58 (%) |
---|---|
Age (range), years | 65 (39–85) |
Sex | |
Male | 54 (93.1) |
Female | 4 (6.9) |
ECOG performance status | |
1 | 53 (91.4) |
≥ 2 | 5 (8.6) |
Histologic type | |
Squamous cell carcinoma | 58 (100.0) |
Alcohol consumption | |
Ever | 48 (82.8) |
Never | 10 (17.2) |
Smoking | |
Ever | 52 (89.7) |
Never | 6 (10.3) |
Previous treatment | |
Curative surgery | 26 (44.8) |
Concurrent chemoradiation | 18 (31.0) |
Number of prior chemotherapies | |
1 | 29 (50.0) |
2 | 22 (37.9) |
≥ 3 | 7 (12.1) |
PD-L1 expression (n = 51) | |
TPS | |
≥ 1% | 16 (31.4) |
≥ 10% | 9 (17.6) |
≥ 20% | 7 (13.7) |
CPS | |
≥ 1 | 29 (56.9) |
≥ 10 | 14 (27.5) |
≥ 20 | 9 (17.6) |
ECOG Eastern Cooperative Oncology Group, PD-L1 programmed cell death ligand-1, TPS tumor proportion score, CPS combined positive score
PD-L1 and CD8 expression
Of the 58 patients included in this study, 51 had samples suitable for the assessment of PD-L1 expression. Based on TPS, 16 (31.4%) patients were determined to be PD-L1-positive (TPS ≥ 1%), including 9 (17.6%) and 7 (13.7%) patients with a TPS ≥ 10% and ≥ 20%, respectively. Based on CPS, 29 (56.9%) patients were determined to be PD-L1-positive (CPS ≥ 1), including 14 (27.5%) and 9 (17.6%) patients with a CPS ≥ 10 and ≥ 20, respectively.
In the CD8 IHC test, 45 and 40 patients had adequate samples for evaluation of IT and PT microscopic fields, respectively: 5 samples were suitable for only IT evaluation, because the representative microscopic fields from tumor samples obtained by small biopsies did not contain an adequate number of PT fields. IT CD8 + T cell infiltration was positive in 11 of 45 (24.4%) samples, and PT CD8 + T cell infiltration was positive in 23 of 40 (57.5%) samples.
Efficacy of nivolumab and predictive role of PD-L1 or CD8 expression
A total of 57 patients, excluding one with no measurable lesion at the baseline image, were included in the response analysis. Among them, six patients were lost to a follow-up imaging evaluation due to the aggravation of cancer-related symptoms. Although their diseases were most probably suspected to be non-responsive to nivolumab, they were included in the analysis of ORR as being an unevaluable objective response. Finally, 11 of 57 patients achieved a partial response with an ORR of 19.3% (11/57) (Table 2). Eleven (19.3%) additional patients achieved stable disease.
Table 2.
N = 58 (%) | |
---|---|
Best response (n = 57)a | |
Partial response | 11 (19.3) |
Stable disease | 11 (19.3) |
Progressive disease | 29 (50.9) |
Not evaluable | 6 (10.5) |
Objective response rate | 11 (19.3) |
Median duration of treatment (range), months | 1.9 (0.5–25.4) |
Median time to response (range), months | 1.6 (1.0–3.2) |
Median duration of response (range), months | 6.5 (4.1–22.4) |
Discontinued treatment | 47 (81.0) |
Reason for treatment discontinuation (n = 47)b | |
Disease progression | 39 (83.0) |
Symptomatic deterioration | 4 (8.5) |
Transfer to another hospital | 1 (2.1) |
Follow-up loss | 3 (6.4) |
aOne patient with only non-measurable lesions, who showed non-CR/non-PD, was excluded from the response analysis
bOf 48 patients who showed a disease progression according to RECIST v1.1, one patient maintained clinical benefit and continued with nivolumab treatment
Although not statistically significant due to the small number of patients analyzed, patients with higher PD-L1 expression levels (1%, 10%, and 20% on TPS and 1, 10, and 20 on CPS) tended to show a higher response rate than those with lower PD-L1 expression (supplementary Table 1; N = 51 for patients with PD-L1 tested). However, even patients with PD-L1-negative tumors according to CPS, demonstrated an ORR of 18.2% (4 of 22). The waterfall plot (Fig. 1; N = 49 patients with baseline measurable lesions and at least one imaging evaluation after treatment initiation) additionally showed a consistent response depth regardless of PD-L1 positivity analyzed according to CPS.
The median duration of therapy for the total study population (N = 58) was 1.9 months (range 0.5–25.4). For 11 patients with a partial response, the median time to initial response was 1.6 months (range 1.0–3.2) and the median duration of the response was 6.5 months (range 4.1–22.4). The swimmer plot (N = 58) in Fig. 2 shows that patients demonstrated a durable response irrespective of PD-L1 CPS positivity. One patient, whose tumor expressed PD-L1 CPS 5 and TPS 0%, showed a dramatic tumor shrinkage maintained over two years (Fig. 3 the patient represented by the bar ranked highest in Fig. 2).
The median follow-up duration for overall survival was 13.8 months. For the entire study population (N = 58), the median PFS was 2.1 months (95% CI 1.8–2.3) and the median overall survival was 7.4 months (95% CI 4.8–10.0) (supplementary Fig. 3). The comparison of survival outcomes according to PD-L1 expression status indicated a trend, albeit not significant, toward better PFS in patients with a PD-L1 CPS ≥ 1 (2.4 [95% CI 1.4–3.4] vs. 1.6 [0.8–2.4], P = 0.20) (Fig. 4a), a CPS ≥ 10 (2.6 [1.1–4.2] vs. 2.0 [1.5–2.6] months, P = 0.24) (Fig. 4b), a TPS ≥ 10% (3.3 [1.3–5.4] vs. 2.0 [1.5–2.6] months, P = 0.15) (Fig. 4d), and a TPS ≥ 20% (7.5 [0.0–15.9] vs. 2.0 [1.6–2.5] months, P = 0.069) (Fig. 4f). A significant difference in PFS was seen among patients with a CPS ≥ 20 (7.5 [1.8–13.1] vs. 1.9 [1.4–2.3] months, P = 0.05) (Fig. 4c). The 12-month PFS rate was 38% for those with a CPS ≥ 20 and 11% for those with a CPS < 20 (Fig. 4c); and 38% for those with a TPS ≥ 20% and 10% for those with a TPS < 10% (Fig. 4f).
Response rates and survival outcomes did not differ significantly according to CD8 + T cell infiltration (supplementary Table 2, supplementary Fig. 5). However, patients with a PD-L1 CPS ≥ 10 or positive for IT CD8 + T cell infiltration showed a numerically longer survival duration (PFS: 2.4 vs. 1.7 months, P = 0.34; OS: 7.5 vs. 5.5, P = 0.41) and nearly two-fold ORR (24.0 vs. 10.5%) and disease control rate (44.0 vs. 21.1%) compared to those whose tumor was negative for both markers (Fig. 5).
Safety of nivolumab
There were 11 treatment-related AEs in 10 patients: 8 cases of hypothyroidism (grade 2), 2 cases of pneumonitis (two grade 1), and 1 colitis (grade 3). Treatment discontinuation was required for only one patient who developed grade 3 colitis. Two patients with grade 1 pneumonitis presented no symptoms but were diagnosed by ground-glass infiltrates on scheduled imaging evaluation, while maintaining a partial remission status. Both patients continued with nivolumab treatment and were closely followed-up for any symptom or imaging aggravation.
Discussion
In this study, we confirmed the clinical efficacy of nivolumab as second- or further-line therapy for ESCC from real-world data. The efficacy values from our study are compatible with those from previous prospective studies [8–10]. The ORR of 19.3% that we obtained is very similar to that of 17.2% or 21.9% (centrally or investigator-assessed, respectively) from ATTRACTION-1, 16.7% from the ESCC subgroup of KEYNOTE-181, and 19.3% from ATTRACTION-3. However, the median overall survival of 7.4 months was slightly shorter than those from prior prospective trials ranging from 8.2 to 10.9 months.
Many factors contribute to the relatively shorter duration of overall survival in our study. First, five patients (9%, 5 of 58) who had ECOG PS 2 or 3 when they first received nivolumab were included in our study, reflecting the patient subgroup encountered in the real-world clinic. These five patients showed a very short overall survival duration of 3.2 months. Poor PS has been previously reported as a negative predictive marker for survival outcome of immunotherapy [14]. In comparison to our study, the prior three prospective studies included only patients with a good PS of 0 or 1 [8–10].
Second, only half of our study population received nivolumab as second-line therapy, while the other half received nivolumab as third-, fourth- or fifth-line therapy. In comparison, all patients enrolled in ATTRACTION-3 and KEYNOTE-181 studies were administered nivolumab and pembrolizumab, respectively, as second-line therapy only [8, 10]. In the subgroup analysis not presented in our Results section, the median overall survival differed according to line of therapy: 7.8 months for patients who received nivolumab as second-line therapy, 6.0 months for those receiving it as third-line therapy, and 3.2 months for those receiving it as fourth- or fifth-line therapy (P = 0.011).
Two phase III trials showed inconclusive results regarding the predictive role of PD-L1 expression in the efficacy of PD-1 inhibitors [8, 10]. Although the whole study population in KEYNOTE-181 with PD-L1 CPS ≥ 10, which consisted of ESCC and EAC, showed a superior overall survival when treated with pembrolizumab compared to chemotherapy, this survival benefit was not statistically significant for the SQCC subgroup [8]. On the other hand, while the ATTRACTION-3 study demonstrated nivolumab treatment reduced the risk of death in patients with PD-L1 TPS ≥ 1%, the actual overall survival duration did not differ between the two groups. Furthermore, there was no significant interaction between nivolumab treatment efficacy and the status of PD-L1 expression [10]. However, the ATTRACTION-3 did not report clinical outcomes according to CPS or other cutoff values by TPS.
To determine whether the efficacy of nivolumab is more predictable by CPS or other cutoff values by TPS, we measured the expression of PD-L1 using both TPS and CPS with various cut-off levels. Our data suggest that a CPS cutoff value of 20 predicts a significant PFS benefit of nivolumab treatment, while a CPS/TPS cutoff value of 10% was more predictable for treatment benefit compared to the cutoff value of 1%. However, the differentiating power of PD-L1 expression was minimal for overall survival.
Previous PD-1/PD-L1 trials across many tumor types demonstrated that the predictive role of PD-L1 expression, if any, is more prominent for overall survival than for PFS [15–17]. In our study, however, PD-L1 expression failed to show any clinically meaningful difference in overall survival. This lack of prediction may be attributable to the large proportion of PD-L1-negative patients in our study population who received post-nivolumab chemotherapy. Fifty-five percent of patients (26 of 47 patients who stopped nivolumab) in our study received post-nivolumab chemotherapy, which was a similar proportion to that reported in the ATTRACTION-3 study [10]. However, when we analyzed these patients according to PD-L1 status, 45% (10/22) belonged to PD-L1-positive, 68% (13/19) to PD-L1-negative, and 50% (3/6) to the PD-L1-unknown group by CPS. Prevalent use of post-nivolumab chemotherapy in the PD-L1-negative group may have improved post-progression survival, leading to attenuation of the predictive effect of PD-L1 expression.
Additionally, we evaluated the role of CD8 + T cell infiltration as a predictive or prognostic marker for nivolumab treatment in ESCC. CD8 + T cells are an important component of tumor-infiltrating lymphocytes, which manifest as a host immune response against cancer cells. However, there are several contrasting reports on the prognostic or predictive roles of CD8 + T cells [18, 19]. Our study showed that infiltration of CD8 + T cells can be used adjunctively with PD-L1 expression as a predictive marker. Though this issue should be further investigated, the lack of predictive power of CD8 + T cell infiltration alone is supported by a previous study, which reported that the expression of human leukocyte antigen (HLA) class I, a prerequisite molecule for activating CD8 + T cells, is deficient in ESCC, suggesting that some of these CD8 + T cells are inactive in the anti-tumor immune response [20].
Biomarker analysis using PD-L1 and CD8 + T cells demonstrated that PD-L1 expression with or without supplementary CD8 + T cell infiltration has a predictive role for nivolumab therapy. However, the predictive power was not sufficient to exclude nivolumab as a treatment option for patients with weak or negative PD-L1 expression or CD8 + T cell infiltration. Further search is needed to identify novel and reliable biomarkers.
There are several limitations to this study. Due to its retrospective nature, our study included a heterogeneous population with regard to PS and treatment line. Therefore, it was difficult to evaluate the long-term effect of nivolumab on overall survival, although efficacy regarding response rate and PFS, which require a relatively shorter duration of follow-up, was sufficiently analyzed. On the other hand, our use of real-world data represents the patient population encountered in the clinic and provides evidence for the role of nivolumab as second- or further-line therapy for patients with advanced ESCC.
In conclusion, our study suggests that nivolumab is a promising option in subsequent treatment lines, with an acceptable toxicity profile for patients with advanced ESCC. This drug should be considered for ESCC patients who experience progression after first- or second-line chemotherapy.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Author contribution
JL, YLC, and JMS contributed to the study conception and design. Material preparation, data collection and analysis were performed by JL, BK, HAJ, YLC, and JMS. The first draft of the manuscript was written by JL and JMS, and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.
Funding
This study was supported by the National R & D Program for Cancer Control, Ministry of Health & Welfare, Korea [1720180].
Availability of data and material
The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.
Code availability
Not applicable.
Compliance with ethical standards
Conflict of interest
The authors have no conflict of interest to declare.
Ethics approval
This study was approved by the Institutional Review Board of Samsung Medical Center with a waiver for informed consent in view of the retrospective nature of the study and all the procedures being performed were part of the routine care (IRB No. 2020-03-087).
Consent to participate
Not applicable.
Consent to publish
Not applicable.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Jiyun Lee, Binnari Kim contributed equally to this work.
References
- 1.Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- 2.Arnold M, Laversanne M, Brown LM, et al. Predicting the future burden of esophageal cancer by histological subtype: international trends in incidence up to 2030. Am J Gastroenterol. 2017;112(8):1247–1255. doi: 10.1038/ajg.2017.155. [DOI] [PubMed] [Google Scholar]
- 3.Cancer.go.kr (2013) National cancer information center. https://www.cancer.go.kr/lay1/program/S1T211C223/cancer/view.do?cancer_seq=4277. Accessed 06 April 2020
- 4.Lin Y, Totsuka Y, He Y, et al. Epidemiology of esophageal cancer in Japan and China. J Epidemiol. 2013;23(4):233–242. doi: 10.2188/jea.je20120162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kim M, Keam B, Kim TM, et al. Phase II study of irinotecan and cisplatin combination chemotherapy in metastatic unresectable esophageal cancer. Cancer Res Treat. 2017;49(2):416–422. doi: 10.4143/crt.2016.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Robert C, Schachter J, Long GV, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372(26):2521–2532. doi: 10.1056/NEJMoa1503093. [DOI] [PubMed] [Google Scholar]
- 7.Zitvogel L, Kroemer G. Targeting PD-1/PD-L1 interactions for cancer immunotherapy. Oncoimmunology. 2012;1(8):1223–1225. doi: 10.4161/onci.21335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Kojima T, Muro K, Francois E, et al. Pembrolizumab versus chemotherapy as second-line therapy for advanced esophageal cancer: Phase III KEYNOTE-181 study. J Clin Oncol. 2019;37(4_suppl):2–2. doi: 10.1200/JCO.2019.37.4_suppl.2. [DOI] [Google Scholar]
- 9.Kudo T, Hamamoto Y, Kato K, et al. Nivolumab treatment for oesophageal squamous-cell carcinoma: an open-label, multicentre, phase 2 trial. Lancet Oncol. 2017;18(5):631–639. doi: 10.1016/s1470-2045(17)30181-x. [DOI] [PubMed] [Google Scholar]
- 10.Kato K, Cho BC, Takahashi M, et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20(11):1506–1517. doi: 10.1016/s1470-2045(19)30626-6. [DOI] [PubMed] [Google Scholar]
- 11.Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45(2):228–247. doi: 10.1016/j.ejca.2008.10.026. [DOI] [PubMed] [Google Scholar]
- 12.Kulangara K, Zhang N, Corigliano E, et al. Clinical utility of the combined positive score for programmed death ligand-1 expression and the approval of pembrolizumab for treatment of gastric cancer. Arch Pathol Lab Med. 2019;143(3):330–337. doi: 10.5858/arpa.2018-0043-OA. [DOI] [PubMed] [Google Scholar]
- 13.Schumacher K, Haensch W, Roefzaad C, et al. Prognostic significance of activated CD8(+) T cell infiltrations within esophageal carcinomas. Cancer Res. 2001;61(10):3932–3936. [PubMed] [Google Scholar]
- 14.Cho JH, Jung HA, Lee SH, et al. Impact of EGFR mutation on the clinical efficacy of PD-1 inhibitors in patients with pulmonary adenocarcinoma. J Cancer Res Clin Oncol. 2019;145(5):1341–1349. doi: 10.1007/s00432-019-02889-0. [DOI] [PubMed] [Google Scholar]
- 15.Cohen EEW, Soulieres D, Le Tourneau C, et al. Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): a randomised, open-label, phase 3 study. Lancet. 2019;393(10167):156–167. doi: 10.1016/s0140-6736(18)31999-8. [DOI] [PubMed] [Google Scholar]
- 16.Mok TSK, Wu YL, Kudaba I, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet. 2019;393(10183):1819–1830. doi: 10.1016/s0140-6736(18)32409-7. [DOI] [PubMed] [Google Scholar]
- 17.Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389(10066):255–265. doi: 10.1016/s0140-6736(16)32517-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Erdag G, Schaefer JT, Smolkin ME, et al. Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Can Res. 2012;72(5):1070–1080. doi: 10.1158/0008-5472.CAN-11-3218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bromwich EJ, McArdle PA, Canna K, et al. The relationship between T-lymphocyte infiltration, stage, tumour grade and survival in patients undergoing curative surgery for renal cell cancer. Br J Cancer. 2003;89(10):1906–1908. doi: 10.1038/sj.bjc.6601400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hosch SB, Izbicki JR, Pichlmeier U, et al. Expression and prognostic significance of immunoregulatory molecules in esophageal cancer. Int J Cancer. 1997;74(6):582–587. doi: 10.1002/(sici)1097-0215(19971219)74:6<582::aid-ijc4>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.
Not applicable.