Skip to main content
Erschienen in: BMC Cancer 1/2019

Open Access 01.12.2019 | Research article

Fluoropyrimidine with or without platinum as first-line chemotherapy in patients with advanced gastric cancer and severe peritoneal metastasis: a multicenter retrospective study

verfasst von: Hiroyuki Arai, Satoru Iwasa, Narikazu Boku, Masahiro Kawahira, Hirofumi Yasui, Toshiki Masuishi, Kei Muro, Keiko Minashi, Shuichi Hironaka, Naoki Fukuda, Daisuke Takahari, Takako Eguchi Nakajima

Erschienen in: BMC Cancer | Ausgabe 1/2019

Abstract

Background

There is no standard first-line chemotherapy for advanced gastric cancer with severe peritoneal metastasis. Although fluoropyrimidine is often used, its efficacy is limited, and it remains unclear whether combination therapy with platinum improves clinical outcomes.

Methods

This retrospective study involved patients at six Japanese academic hospitals between 2010 and 2016. Patients with advanced gastric cancer and severe peritoneal metastasis were included if they had massive ascites and/or inadequate oral intake requiring intravenous nutritional support. We then compared the efficacy and safety of fluoropyrimidine monotherapy with those of fluoropyrimidine/platinum combination therapy.

Results

Compared with the combination therapy group (n = 64), the monotherapy group (n = 65) had worse general health (more patients with elderly age, performance status > 2, and having both massive ascites and inadequate oral intake). Both overall survival (9.0 vs 5.0 months, p < 0.01) and progression-free survival (4.3 vs 2.3 months, p < 0.01) were significantly longer in the combination group, and the significance remained after adjusting for prognostic variables (hazard ratios of 0.47 and 0.41, respectively; p < 0.01). Improvements in ascites and oral intake were also greater in the combination group. Although neutropenia (grade ≥ 3) occurred more frequently with combination therapy, both treatments in this study were tolerable.

Conclusions

Combination therapy with fluoropyrimidine and platinum might be more effective than monotherapy with fluoropyrimidine and was tolerable for patients with advanced gastric cancer and severe peritoneal metastasis.
Abkürzungen
AGC
Advanced gastric cancer
OS
Overall survival
PFS
Progression-free survival
SPM
Severe peritoneal metastasis
TTF
Time to treatment failure

Background

Gastric cancer (GC) is the fifth most common cancer and the third leading cause of cancer-related death worldwide [1]. The standard first-line chemotherapy for patients with erb-b2 receptor tyrosine kinase 2 (ERBB2, also known as HER2)-negative advanced GC (AGC) in Japan is a combination therapy with fluoropyrimidine plus platinum (e.g., S-1 or capecitabine plus cisplatin or oxaliplatin) [24].
Peritoneal metastasis is very common in AGC and causes a range of serious clinical complications, including massive ascites, bowel obstruction, jaundice, and hydronephrosis, that worsen prognosis and quality of life [5, 6]. When massive ascites is present, intolerable abdominal fullness often makes it difficult to administer cisplatin which requires adequate hydration. Subacute or acute bowel obstruction causing nausea, vomiting, and malabsorption, also makes stable oral fluoropyrimidine intake impractical. For these reasons, patients with severe peritoneal metastasis (SPM) associated with massive ascites and/or inadequate oral intake were excluded from pivotal phase III trials [24, 7]. Even in a phase III trial (Japan Clinical Oncology Group [JCOG] 0106), comparing fluorouracil (5-FU) by continuous infusion (ci) with 5-FU plus methotrexate (MTX) in AGC patients with peritoneal metastasis, patients with massive ascites were excluded and only a few patients (14% [32/237]) with inadequate oral intake were included [8]. Based on such backgrounds, no standard first-line chemotherapy has been established and an unmet need exists for prolongation the survival time in AGC patients with SPM. On the other hand, adaptation of patients to chemotherapy should be carefully judged before treatment initiation because an aggressive disease course can immediately change the condition of patients into a non-adaptive state to chemotherapy.
In clinical practice, fluoropyrimidine monotherapy regimens, such as options including 5-FU plus l-leucovorin (l-LV), 5-FU ci, and 5-FU plus MTX, are generally used as first-line chemotherapy for patients with AGC and SPM [9, 10]. However, retrospective studies have shown that fluoropyrimidine monotherapy has modest efficacy and feasible toxicity with median survival times of only 4.6–6.0 months, indicating that further investigation is warranted [9, 10]. One option is to use platinum in combination with fluoropyrimidine [11]. Here, oxaliplatin is a third-generation platinum agent and suitable even for patients with massive ascites because hydration is needless. Recently, oxaliplatin was approved for the treatment of GC in Japan based on its non-inferiority to cisplatin in phase III trials of patients with AGC without SPM [4, 12].
The lack of prospective data in patients with AGC and SPM makes it unclear whether combination therapy with fluoropyrimidine plus platinum could be more effective than fluoropyrimidine monotherapy in patients with AGC and SPM. Therefore, we investigated the efficacy and safety of fluoropyrimidine/platinum combination therapy compared with fluoropyrimidine monotherapy when used as the first-line chemotherapy in patients with AGC and SPM.

Methods

Study design

This was a retrospective study of patients with AGC and SPM who received first-line chemotherapy between July 2010 and September 2016 at six institutions in Japan. We compared the efficacy and safety of fluoropyrimidine/platinum combination therapy (the FP group) with those of fluoropyrimidine monotherapy (the F group). All patient data were extracted from a database at each center. This study was approved by the institutional review board in each center. Written informed consent was obtained from all patients before treatment initiation.

Eligibility criteria and definitions

All the patients in this study had SPM. SPM was defined as peritoneal metastasis associated with massive ascites and/or inadequate oral intake, with the latter defined as requiring intravenous nutritional support. The degree of ascites was evaluated by computed tomography and classified as follows: “none” if undetectable; “mild” if localized to the pelvic cavity or upper abdominal cavity; “moderate” if inconsistent with either mild or massive ascites; and “massive” if extending continuously between the pelvic cavity and upper abdominal cavity. The eligibility criteria for this study were as follows: (1) histologically proven adenocarcinoma of the stomach or gastroesophageal junction; (2) HER2-negative or unknown tumor; (3) having SPM; (4) absence of concomitant advanced malignant disease; and (5) if the patient had recurrent disease, the recurrence occurred at least 6 months after the last dose of adjuvant chemotherapy. We excluded patients who had participated in the randomized phase II/III trial of 5-FU/l-LV compared with 5-FU/l-LV plus paclitaxel for AGC with SPM (JCOG1108/WJOG7312G trial: UMIN000010949) because the trial was ongoing and not available for analysis at start of this study. We also excluded patients who had inadequate hepatic and/or renal function (serum total bilirubin > 3.0 mg/dl, serum creatinine > 2.0 mg/dl).

Chemotherapy

The F group received any fluoropyrimidine monotherapy regimen with or without chemical modulators (e.g., l-LV or MTX). The FP group received cisplatin or oxaliplatin in combination with any fluoropyrimidine. If a 5-FU regimen was modified to an S-1 regimen due to improved oral intake, the S-1 regimen was regarded as second-line chemotherapy.

Assessment of response in ascites and improvement of Oral intake

We compared the degree of ascites between baseline and during treatment, and determined the best response in ascites as follows: “complete response (CR)” if the ascites completely disappeared; “partial response (PR)” if there was a decrease by at least one degree from baseline; “stable disease” if there was no change from baseline; “progressive disease” if there was an increase by at least one degree from baseline; and “not evaluated” if it was impossible to evaluate because ascites was drained before assessment or because there was no recorded assessment. We defined the response rate in ascites as the proportion of patients with the best CR or PR among those with ascites at baseline.
The improvement rate of oral intake was also defined as the proportion of patients whose oral intake recovered and did not require nutritional support for at least 7 days among the patients who had inadequate oral intake at baseline.

Statistical analysis

We compared patient demographics between the FP and F groups. P values were estimated by chi-square tests for categorical variables and by Mann–Whitney U tests for continuous variables. Time to treatment failure (TTF) was defined as the time from treatment start to the last dose of chemotherapy. Overall survival (OS) was defined as the time from treatment start to death from any cause, and progression-free survival (PFS) was defined as the time from treatment start to disease progression or death from any cause. Patients who had no events during the observation period were censored at the last follow-up date (cut-off date, June 30, 2017).
Both OS and PFS were estimated by the Kaplan–Meier method and compared between the groups by the log-rank test. Hazard ratios (HRs) and confidence intervals (CIs) were then estimated using Cox proportional hazards models in both univariate and multivariate analyses. Covariates with a p-value < 0.20 in the univariate analysis were included into the multivariate analysis, and the Eastern Cooperative Oncology Group performance status (PS; i.e., 0–1 or 2–4) and SPM subtype (i.e., massive ascites, inadequate oral intake, or both) were also included because these covariates were considered clinically important. The response rate in ascites, the improvement rate of oral intake, and the safety of each regimen were compared by chi-square tests.
We also identified patients who might not be adaptive to any type of chemotherapy to prevent such patients from undergoing chemotherapy as they would not benefit from it. To this end, we considered patients who died within 90 days after treatment initiation to be potentially “non-adaptive” and survivors to be “adaptive.” Further, we compared characteristics between the “non-adaptive” and “adaptive” patients and conducted univariate analysis to estimate odds ratio (OR) of each risk factor for early death, i.e., death within 90 days after treatment initiation, by logistic regression. In this analysis, we used the median value of all patients as the cut-off value, e.g., serum albumin (3.1 g/ml) and CRP (2.2 mg/dl) levels.
All analyses were two-sided, and a p-value < 0.05 was considered statistically significant. All statistical analyses were performed using StatView ver 5.0 software (SAS Institute, Cary, NC, USA).

Results

Patients

After excluding 10 ineligible patients (6 participated in JCOG1108/WJOG7312G trial, 3 had inadequate hepatic function, and 1 had inadequate renal function) from the available cohort, we finally recruited 129 patients (64 in the FP group and 65 in the F group). The baseline characteristics of the participants are shown in Table 1. Overall, 40% had a PS > 2 (including 7 patients with PS 3 and 1 patient with PS 4), and the median (range) serum albumin level was 3.1 (1.8–4.4) g/ml. The F group tended to be older (median, 67 vs 62 years) and include more patients with a PS > 2 (51% vs 30%), fewer patients with massive ascites only (35% vs 61%), and more patients with both massive ascites and inadequate oral intake (35% vs 14%). Median serum albumin and C-reactive protein levels were comparable between both groups.
Table 1
Patient characteristics
Characteristics
All patients
FP group
F group
p-value
(n = 129)
(n = 64)
(n = 65)
No. (%)
No. (%)
No. (%)
Age (years)
 Median (range)
65 (24–94)
62 (27–94)
67 (24–89)
0.02
 < 65
61 (47)
36 (56)
25 (38)
0.04
> 65
68 (53)
28 (44)
40 (62)
 
Sex
 Male
68 (53)
33 (52)
35 (54)
0.80
 Female
61 (47)
31 (48)
30 (46)
 
ECOG PS
 0
9 (7)
4 (6)
5 (8)
 
 1
68 (53)
41 (64)
27 (42)
 
 2
44 (34)
16 (25)
28 (43)
 
 3
7 (5)
2 (3)
5 (8)
 
 4
1 (1)
1 (2)
0 (0)
 
 0–1
77 (60)
45 (70)
32 (49)
0.01
 2–4
52 (40)
19 (30)
33 (51)
 
Histology
 Intestinal
22 (17)
10 (16)
12 (18)
0.81
 Diffuse
102 (79)
52 (81)
50 (77)
 
 Unknown
5 (4)
2 (3)
3 (5)
 
Disease status
 Advanced
120 (93)
58 (91)
62 (95)
0.29
 Recurrent
9 (7)
6 (9)
3 (5)
 
Primary tumor
 Presence
113 (88)
54 (84)
59 (91)
0.27
 Absence
16 (12)
10 (16)
6 (9)
 
Primary site
 Stomach
123 (95)
59 (92)
64 (98)
0.09
 GEJ
6 (5)
5 (8)
1 (2)
 
No. of metastatic sites
 1–2
100 (78)
49 (77)
51 (78)
0.80
> 3
29 (22)
15 (23)
14 (22)
 
Target lesion
 Presence
56 (43)
24 (38)
32 (49)
0.18
 Absence
73 (57)
40 (62)
33 (51)
 
Subtype of SPM
 Massive ascites
62 (48)
39 (61)
23 (35)
0.01
 Inadequate oral intake
35 (27)
16 (25)
19 (29)
 
 Both
32 (25)
9 (14)
23 (35)
 
Serum albumin level (g/ml)
 Median (range)
3.1 (1.8–4.4)
3.1 (1.8–4.3)
3.1 (1.8–4.4)
0.87
Serum CRP level (mg/dl)
 Median (range)
2.2 (0.0–24.0)
1.8 (0.1–20.7)
2.6 (0.0–24.0)
0.86
CRP C-reactive protein, ECOG PS: Eastern Cooperative Oncology Group performance status, GEJ gastroesophageal junction, SPM severe peritoneal metastasis

Chemotherapy exposure

S-1 plus cisplatin (n = 27), modified FOLFOX6 (n = 17), S-1 plus oxaliplatin (n = 14), and 5-FU plus cisplatin (n = 6) were used in the FP group. By contrast, 5-FU/l-LV (n = 39), S-1 (n = 22), 5-FU ci (n = 3), and 5-FU/MTX (n = 1) were used in the F group (Fig. 1). The dosage and treatment schedules of each regimen were similar to those previously reported [2, 4, 8, 13, 14].
The first-line treatment was discontinued in all patients at median TTFs of 3.3 and 1.4 months in the FP and F groups, respectively. Second-line chemotherapy was given to 46 patients (72%) in the FP group and 33 patients (51%) in the F group (p = 0.01). Most patients in both the FP group (33/46; 72%) and the F group (25/33; 76%) received taxane-based chemotherapy.

Reasons for discontinuation

Reasons for treatment discontinuation in the FP group were disease progression (78% [50/64]), adverse events (6% [4/64]), change from 5-FU to S-1 regimens because of improved oral intake (2% [1/64]), withdrawal of consent (2% [1/64]), and other reasons (13% [8/64]); the adverse events leading to treatment discontinuation in the FP group were peripheral neuropathy (n = 2), fatigue (n = 1), and stomatitis (n = 1). Reasons for treatment discontinuation in the F group were disease progression (77% [50/65]), adverse events (9% [6/65]), change from 5-FU to S-1 regimens because of improved oral intake (11% [7/65]), and other reasons (3% [2/65]); the adverse events leading to treatment discontinuation in the F group were sepsis (n = 2), febrile neutropenia (n = 1), appetite loss (n = 1), stomatitis (n = 1), and angina (n = 1).

Efficacy

The number of OS events was 50 (78%) in the FP group and 61 (94%) in the F group. Totally, all the patients showed median OS and PFS of 6.7 and 3.1 months, respectively. Analysis by SPM subtypes revealed OS for patients with massive ascites only, inadequate oral intake only, and both of 8.0, 9.0, and 5.4 months, respectively, with corresponding PFS times of 2.8, 4.0, and 3.4 months, respectively (Fig. 2).
The median OS was 9.0 months in the FP group and 5.0 months in the F group, with a statistical significance (HR, 0.56; 95% CI, 0.39–0.82; log-rank p < 0.01) (Fig. 3). Univariate analysis revealed that intestinal histological type, recurrent disease, ≤ 2 metastatic sites, and serum albumin ≥3.1 g/ml were predictors of better OS (p < 0.20). After adjustment for these factors, PS and the SPM subtype, OS in the FP group remained superior to that in the F group (HR, 0.47; 95% CI, 0.31–0.72; p < 0.01) (Table 2).
Table 2
Univariate and multivariate analyses of the prognostic factors of OS in AGC patients with SPM (n = 129)
Variables
Univariate analysis
Multivariate analysis
HR
[95% CI]
p-value
HR
[95% CI]
p-value
Regimen
 FP (vs. F)
0.56
[0.39–0.82]
< 0.01
0.47
[0.31–0.72]
< 0.01
Age
 ≥65 (vs. < 65)
0.80
[0.55–1.17]
0.25
   
Sex
 Female (vs. male)
1.05
[0.72–1.54]
0.78
   
PS
 2–4 (vs. 0–1)
1.22
[0.83–1.79]
0.32
0.95
[0.62–1.48]
0.83
Histology
 Intestinal (vs. diffuse)
0.71
[0.43–1.19]
0.19
0.70
[0.41–1.19]
0.18
 Unknown (vs. diffuse)
NA
 
NA
NA
 
NA
Disease status
 Recurrent (vs. advanced)
0.58
[0.25–1.32]
0.19
0.86
[0.36–2.10]
0.75
Primary site
 GEJ (vs. stomach)
0.98
[0.40–2.41]
0.96
   
No. of metastatic sites
 ≥3 (vs. 1–2)
1.43
[0.92–2.23]
0.11
1.64
[0.97–2.76]
0.07
Target lesion
 Absence (vs. presence)
0.95
[0.65–1.38]
0.78
   
Subtype of SPM
 Inadequate oral intake (vs. massive ascites)
0.81
[0.52–1.29]
0.38
0.58
[0.33–1.00]
0.05
 Both (vs. massive ascites)
1.09
[0.69–1.71]
0.71
0.76
[0.44–1.29]
0.31
Serum albumin level
 < 3.1 g/ml (vs. > 3.1 g/ml)
1.42
[0.97–2.07]
0.07
1.43
[0.94–2.17]
0.09
AGC advanced gastric cancer, CI confidence interval, ECOG PS Eastern Cooperative Oncology Group performance status, GEJ gastroesophageal junction, HR hazard ratio, NA not assessed, OS overall survival, SPM severe peritoneal metastasis
PFS was also significantly longer in the FP group than in the F group (median, 4.3 vs 2.3 months; HR, 0.44; 95% CI, 0.30–0.66; log-rank p < 0.01) (Fig. 3). Univariate analysis identified the recurrent disease and having only inadequate oral intake as predictors of better PFS (p < 0.20). After adjustment for these factors and PS, PFS in the FP group remained superior to that in the F group (HR, 0.41; 95% CI, 0.26–0.64; p < 0.01) (Table 3).
Table 3
Univariate and multivariate analyses of the prognostic factors of PFS in AGC patients with SPM (n = 129)
Variables
Univariate analysis
Multivariate analysis
HR
[95% CI]
p-value
HR
[95% CI]
p-value
Regimen
 FP (vs. F)
0.44
[0.30–0.66]
< 0.01
0.41
[0.26–0.64]
< 0.01
Age
 ≥65 (vs. < 65)
0.99
[0.67–1.45]
0.95
   
Sex
 Female (vs. male)
0.96
[0.65–1.41]
0.82
   
ECOG PS
 2–4 (vs. 0–1)
1.28
[0.86–1.90]
0.22
1.04
[0.68–1.60]
0.86
Histology
 Intestinal (vs. diffuse)
0.81
[0.48–1.39]
0.45
   
 Unknown (vs. diffuse)
NA
 
NA
   
Disease status
 Recurrent (vs. advanced)
0.48
[0.20–1.19]
0.11
0.60
[0.24–1.50]
0.27
Primary site
 GEJ (vs. stomach)
0.55
[0.20–1.53]
0.25
   
No. of metastatic sites
 ≥3 (vs. 1–2)
1.28
[0.86–1.90]
0.22
   
Target lesion
 Absence (vs. presence)
1.03
[0.69–1.53]
0.90
   
Subtype of SPM
 Inadequate oral intake (vs. massive ascites)
0.66
[0.41–1.07]
0.09
0.64
[0.40–1.04]
0.07
 Both (vs. massive ascites)
0.88
[0.54–1.44]
0.61
0.57
[0.34–0.96]
0.03
Serum albumin level
 < 3.1 g/ml (vs. > 3.1 g/ml)
1.10
[0.74–1.62]
0.65
   
AGC advanced gastric cancer, CI confidence interval, ECOG PS Eastern Cooperative Oncology Group performance status, GEJ gastroesophageal junction, HR hazard ratio, NA not assessed, PFS progression-free survival, SPM severe peritoneal metastasis
Response rate in ascites was 51% (30/59) in the FP group and 17% (10/60) in the F group (p < 0.01). Improvement rate of oral intake was 64% (16/25) in the FP group and 43% (18/42) in the F group (p = 0.09).

Safety

The incidence of all grades of adverse events was significantly higher in the FP group for leukopenia (63% vs 35%), neutropenia (61% vs 26%), anemia (80% vs 57%), thrombocytopenia (58% vs 14%), and sensory neuropathy (38% vs 0%), compared with the F group. Regarding the incidences of grade 3 or 4 adverse events, only neutropenia was significantly higher in the FP group (36% vs 11%), whereas the others were similar (Table 4). There were no treatment-related deaths. However, we observed death within 30 days form the last dose of chemotherapy in 23 patients (18%) totally: 7 (11%) in the FP group and 16 (25%) in the F group. All of these patients were died from disease progression. This included 7 patients (5%) who died after the first administration of chemotherapy: 1 (2%) in the FP group and 6 (9%) in the F group.
Table 4
Adverse Events
Adverse Event
FP (n = 64)
F (n = 65)
p-value
All Gr.
Gr. 3–4
All Gr.
Gr. 3–4
No. (%)
No. (%)
No. (%)
No. (%)
All Gr.
Gr. 3–4
Leukocytopenia
40 (63)
11 (17)
23 (35)
5 (8)
< 0.01
0.10
Neutropenia
39 (61)
23 (36)
17 (26)
7 (11)
< 0.01
< 0.01
Anemia
51 (80)
12 (19)
37 (57)
15 (23)
< 0.01
0.55
Thrombocytopenia
37 (58)
3 (5)
9 (14)
0 (0)
< 0.01
0.08
Febrile neutropenia
3 (5)
3 (5)
3 (5)
3 (5)
0.98
0.98
Anorexia
42 (66)
10 (16)
37 (57)
17 (26)
0.31
0.14
Nausea
40 (63)
3 (5)
34 (52)
1 (2)
0.24
0.30
Vomiting
18 (28)
1 (2)
23 (35)
2 (3)
0.38
0.57
Diarrhea
16 (25)
3 (5)
18 (28)
1 (2)
0.73
0.30
Stomatitis
14 (22)
1 (2)
7 (11)
0 (0)
0.09
0.31
Fatigue
27 (42)
2 (3)
27 (42)
3 (5)
0.94
0.66
Sensory neuropathy
24 (38)
1 (2)
0 (0)
0 (0)
< 0.01
0.31

Risk factors for early death

We observed early death in 28 patients (22% of 129 patients): 7 patients (11%) in the FP group and 21 patients (32%) in the F group. These 28 patients were deemed to be potentially “non-adaptive” to any type of chemotherapy, whereas the remaining 101 patients were deemed to be “adaptive.” “Non-adaptive” patients had significantly higher serum CRP level than “adaptive” patients (Table 5). Univariate analysis showed two significant risk factors for early death: serum albumin level < 3.1 g/ml (OR, 2.43; 95% CI, 1.02–5.78; p = 0.05) and serum CRP level > 2.2 mg/dl (OR, 2.87; 95% CI, 1.16–7.13; p = 0.02) (Table 6).
Table 5
Characteristics of “adaptive” or “non-adaptive” patients
Characteristics
Adaptive patients
Non-adaptive patients
p-value
(n = 101)
(n = 28)
No. (%)
No. (%)
Age (years)
 Median (range)
65 (24–86)
67 (34–94)
0.09
Sex
 Male
51 (50)
17 (61)
0.34
 Female
50 (50)
11 (39)
 
ECOG PS
 0
8 (8)
1 (4)
 
 1
56 (55)
12 (43)
 
 2
32 (32)
12 (43)
 
 3
4 (4)
3 (11)
 
 4
1 (1)
0 (0)
 
 0–1
64 (63)
13 (46)
0.11
 2–4
37 (37)
15 (54)
 
Disease status
 Advanced
92 (91)
28 (100)
0.10
 Recurrent
9 (9)
0 (0)
 
Primary site
 Stomach
97 (96)
26 (93)
0.48
 GEJ
4 (4)
2 (7)
 
No. of metastatic sites
 1–2
81 (80)
19 (68)
0.17
> 3
20 (20)
9 (32)
 
Subtype of SPM
 Massive ascites
46 (45)
16 (57)
0.50
 Inadequate oral intake
28 (27)
7 (25)
 
 Both
27 (27)
5 (18)
 
Serum albumin level (g/ml)
 Median (range)
3.1 (1.8–4.4)
2.9 (2.0–4.2)
0.13
Serum CRP level (mg/dl)
 Median (range)
1.6 (0.0–20.7)
4.8 (0.1–24.0)
< 0.01
Patients who died within 90 days after treatment initiation were defined as “non-adaptive”
Other patients were defined as “adaptive”
CRP C-reactive protein, ECOG PS Eastern Cooperative Oncology Group performance status, GEJ gastroesophageal junction, SPM severe peritoneal metastasis
Table 6
Univariate analysis for risk factors for early death (within 90 days after treatment initiation)
Variable
Univariate analysis
N
OR
[95% CI]
p-value
Age (years)
 < 65
11/61
1.00
  
 ≥65
17/68
1.52
[0.65–3.56]
0.34
Sex
 Male
17/68
1.00
  
 Female
11/61
0.66
[0.28–1.55]
0.34
ECOG PS
 0–1
13/77
1.00
  
 2–4
15/52
2.00
[0.86–4.65]
0.11
Disease status
 Advanced
28/120
NC
 
NC
 Recurrent
0/9
Primary site
 Stomach
26/123
1.00
  
 GEJ (vs. stomach)
2/6
1.87
[0.32–10.76]
0.49
No. of metastatic sites
 1–2
19/100
1.00
  
 ≥3
9/29
1.92
[0.76–4.87]
0.17
Subtype of SPM
 Massive ascites
16/62
1.00
  
 Inadequate oral intake
7/35
0.72
[0.26–1.96]
0.52
 Both
5/32
0.53
[0.18–1.62]
0.27
Serum albumin level
> 3.1 g/ml
10/68
1.00
  
 < 3.1 g/ml
18/61
2.43
[1.02–5.78]
0.05
Serum CRP level
 < 2.2 mg/dl
8/62
1.00
  
> 2.2 mg/dl
20/67
2.87
[1.16–7.13]
0.02
CI confidence interval, CRP C-reactive protein, ECOG PS Eastern Cooperative Oncology Group performance status, GEJ gastroesophageal junction, NC not calculated, OR odds ratio, SPM severe peritoneal metastasis

Discussion

This is the first study to compare the efficacy and safety of fluoropyrimidine with and without platinum as the first-line chemotherapy for AGC with SPM. This retrospective multicenter study suggested that combination therapy with fluoropyrimidine/platinum might be more effective than fluoropyrimidine monotherapy in terms of the OS, the PFS, and the improvement in both ascites and oral intake, associated with acceptable toxicity. Even considering the difference in patient background, multivariate analysis showed that both OS and PFS were significantly better in the FP group compared with the F group. We also determined significant risk factors for early death that are not likely to promote adaptation to chemotherapy among patients.
The general health condition of the subjects was poor in this study, as shown by the high proportion of patients with a high PS (40% had a PS > 2) and low serum albumin level (the median was 3.1 g/ml). This study also revealed treatment bias: monotherapy more likely to be selected for patients with worse general health statuses. Indeed, the F group included more elderly patients, more patients with a PS > 2, and more patients with ‘both’ SPM subtype. However, this mirrors actual clinical practice for the treatment of patients with AGC and SPM.
Of note, the F group showed comparable outcomes to those reported in previous studies. Iwasa et al. reported that the median TTF and OS were 1.9 and 4.6 months among 92 patients with AGC and SPM receiving 5-FU monotherapy regimens, including 44 (48%) with a PS > 2 [9]. Also, Hara et al. reported median PFS and OS of 2.4 and 6.0 months among 30 patients with AGC and SPM receiving 5-FU/I-LV, including 15 (50%) with a PS > 2 [10]. In our study, the median TTF, PFS, and OS were 1.4, 2.3, and 5.0 months, respectively, in the F group. Given that the median OS for patients with AGC receiving best supportive care (BSC) is about 3 months [15, 16], fluoropyrimidine monotherapy appeared to confer a modest survival benefit of about 2–3 months over BSC. In contrast, in the current study, the median OS in the FP group was 9.0 months, which may account for a minimum of 6-month survival benefit over BSC. We demonstrated that fluoropyrimidine/platinum combination therapy significantly prolonged OS and PFS compared with fluoropyrimidine alone, which was confirmed by the adjusted HR. This result is consistent with SPIRITS trial that demonstrated combination of S-1 plus cisplatin was superior to S-1 alone for the patients with general AGC. Furthermore, combination therapy with fluoropyrimidine/platinum induced a greater improvement in ascites and a greater improvement in oral intake. Such combination therapy could, therefore, be a promising treatment option for improving the prognosis and reducing the undesirable symptoms of AGC with SPM. Looking forward, we have planned further investigations to explore the most promising combination therapies based on fluoropyrimidine and platinum. This study and another small retrospective study of patients with AGC and SPM suggest that the FOLFOX regimen might be a good candidate [14].
Although there was a higher incidence of grade 3 or more neutropenia in the FP group, both FP and F were feasible. However, there was a high incidence of early death within 30 days of last administration (11% in the FP group and 25% in the F group), including too early death within 30 days of treatment start (2% in the FP group and 9% in the F group), because of rapid disease progression. This raises the issues of appropriate patient selection for chemotherapy and of the correct timing of switching to BSC, which require further investigation in future studies.
At present, prolongation of the survival of patients with AGC and SPM is highly warranted. Paclitaxel, which has been shown to exert activity on peritoneal metastasis, is another candidate to develop more effective chemotherapy regimens. When used as first-line chemotherapy, a randomized phase III trial (PHOENIX-GC trial) of patients with AGC and peritoneal metastasis showed that combination therapy with intravenous and intraperitoneal paclitaxel plus S-1 lengthened the OS, albeit without statistical significance, compared with S-1 plus cisplatin (HR, 0.72; 95% CI, 0.49–1.04; p = 0.081) [17]. When used as second-line chemotherapy, a randomized phase II trial (JCOG0407 trial) of patients with AGC and peritoneal metastasis refractory to fluoropyrimidine showed that weekly paclitaxel lengthened the PFS compared with the best available 5-FU regimen (HR, 0.58; 95% CI, 0.38–0.88; p = 0.005) [18]. A first-line combination strategy using paclitaxel has been developed for patients with SPM. The FLTAX regimen, which combines 5-FU/l-LV with paclitaxel, has had its recommended dose and feasibility confirmed for patients with AGC and SPM in a phase I trial [19]. Currently, a randomized phase II/III trial (JCOG1108/WJOG7312G trial: UMIN000010949) is ongoing to evaluate its efficacy and safety compared to 5-FU/l-LV, and this study will provide clinically important information. If the preliminary results can be confirmed, we will need to consider which of FP and FLTAX is more effective for the treatment of AGC with SPM.
To the best of our knowledge, few studies have predicted patients who would be adaptive to first-line chemotherapy in this patient group. To identify such patients, we explored risk factors for early death in this study. We determined early death to occur within 90 days after treatment initiation because this period of life expectancy is generally considered extremely short to initiate chemotherapy. We found two significant risk factors for early death: serum albumin level < 3.1 g/ml and CRP level > 2.2 mg/dl. Lower serum albumin level and higher serum CRP level are the possible indicators of poor patient condition and degree of disease aggressiveness. We suggest that these factors indicate extremely short life expectancy for chemotherapy, thus indicating that these patients should receive BSC. Future studies should expand on our exploratory findings.
This study has two key limitations. First, this was a retrospective study, and we could not exclude the possibility that the outcomes were affected by the different patient characteristics between treatment groups, despite adjustment for the most likely factors in the multivariate analysis. Therefore, these results should be carefully interpreted during consideration for incorporation into clinical practice. To resolve this critical issue, prospective studies are necessary. Second, the definition of SPM included heterogeneous subtypes, and it remains unclear whether the same treatment strategy is appropriate for all potential subtypes. Further discussion of this point is required if we are to develop meaningful treatment options for this population in the future.

Conclusions

Fluoropyrimidine and platinum combination therapy might be more effective than fluoropyrimidine monotherapy for AGC patients with SPM. Further investigation is warranted for development of the treatment options in this population.

Acknowledgements

Not applicable.

Conflict of interest

NB reports personal fees from Yakult, grants and personal fees from Taiho, personal fees from Chugai, during the conduct of the study; grants and personal fees from Ono, grants from Bristol Myers-Squibb, personal fees from Eli Lilly, personal fees from Shionogi, personal fees from Merck Serono, outside the submitted work; KM1 reports grants from Ono Pharmaceutical, grants from MSD, grants from Daiichi Sankyo, grants from Kyowa Hakko Kirin, grants from Shionogi Pharmaceutical, grants from Gilead Sciences, personal fees from Chugai Pharmaceutical, personal fees from Taiho Pharmaceutical, personal fees from Takeda Pharmaceutical, personal fees from Merck Serono, personal fees from Eli Lilly, personal fees from Yakult Honsha, outside the submitted work; SH reports personal fees from Taiho Pharmaceutical Co., Ltd., personal fees from Eli Lilly, personal fees from Bristol-Meyers Squibb Co. Ltd., personal fees from Ono Pharmaceutical Co. Ltd., personal fees from Yakult Honsha, personal fees from Daiichi-Sankyo, outside the submitted work; TEN reports grants and personal fees from Merck Serono, grants and personal fees from Taiho Pharm., grants and personal fees from Chugai Pharm, grants and personal fees from Takeda Pharm, grants from Yakult Honsha, grants and personal fees from Eli Lilly Japan, grants from Eisai Pharm, grants and personal fees from Sanofi, grants from Amgen Astellas BioPharma, grants and personal fees from Ono Pharm, grants from AstraZeneca, personal fees from Sawai Pharm., personal fees from Kyowa Hakko-Kirin, personal fees from Bristol-Myers Squibb, personal fees from Bayer Yakuhin, grants and personal fees from Dainippon Sumitomo, personal fees from Maruho Co. Ltd., personal fees from MSD, personal fees from Mochida Pharm, outside the submitted work.

Funding

We had no financial supports for this study.

Availability of data and materials

The data that support the findings of this study are not available publicly. However, the data are available from the corresponding author upon reasonable request.
This retrospective study was approved by Institutional Review Board (Ethics Committee, St. Marianna University School of Medicine) in accordance with the Japanese Ethical Guidelines for Medical and Health Research Involving Human Subjects. Written informed consent was obtained from all patients before treatment initiation.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
2.
Zurück zum Zitat Koizumi W, Narahara H, Hara T, et al. S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer (SPIRITS trial): a phase III trial. Lancet Oncol. 2008;9:215–21.CrossRef Koizumi W, Narahara H, Hara T, et al. S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer (SPIRITS trial): a phase III trial. Lancet Oncol. 2008;9:215–21.CrossRef
3.
Zurück zum Zitat Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687–97.CrossRef Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687–97.CrossRef
4.
Zurück zum Zitat Yamada Y, Higuchi K, Nishikawa K, et al. Phase III study comparing oxaliplatin plus S-1 with cisplatin plus S-1 in chemotherapy-naive patients with advanced gastric cancer. Ann Oncol. 2015;26:141–8.CrossRef Yamada Y, Higuchi K, Nishikawa K, et al. Phase III study comparing oxaliplatin plus S-1 with cisplatin plus S-1 in chemotherapy-naive patients with advanced gastric cancer. Ann Oncol. 2015;26:141–8.CrossRef
5.
Zurück zum Zitat Fujimura T, Ishii K, Oyama K, et al. A new scoring system for peritoneal metastasis in gastric cancer. Gastric Cancer. 2003;6:146–52.CrossRef Fujimura T, Ishii K, Oyama K, et al. A new scoring system for peritoneal metastasis in gastric cancer. Gastric Cancer. 2003;6:146–52.CrossRef
6.
Zurück zum Zitat Wei J, Wu ND, Liu BR. Regional but fatal: intraperitoneal metastasis in gastric cancer. World J Gastroenterol. 2016;22:7478–85.CrossRef Wei J, Wu ND, Liu BR. Regional but fatal: intraperitoneal metastasis in gastric cancer. World J Gastroenterol. 2016;22:7478–85.CrossRef
7.
Zurück zum Zitat Boku N, Yamamoto S, Fukuda H, et al. Fluorouracil versus combination of irinotecan plus cisplatin versus S-1 in metastatic gastric cancer: a randomised phase 3 study. Lancet Oncol. 2009;10:1063–9.CrossRef Boku N, Yamamoto S, Fukuda H, et al. Fluorouracil versus combination of irinotecan plus cisplatin versus S-1 in metastatic gastric cancer: a randomised phase 3 study. Lancet Oncol. 2009;10:1063–9.CrossRef
8.
Zurück zum Zitat Shirao K, Boku N, Yamada Y, et al. Randomized phase III study of 5-fluorouracil continuous infusion vs. sequential methotrexate and 5-fluorouracil therapy in far advanced gastric cancer with peritoneal metastasis (JCOG0106). Jpn J Clin Oncol. 2013;43:972–80.CrossRef Shirao K, Boku N, Yamada Y, et al. Randomized phase III study of 5-fluorouracil continuous infusion vs. sequential methotrexate and 5-fluorouracil therapy in far advanced gastric cancer with peritoneal metastasis (JCOG0106). Jpn J Clin Oncol. 2013;43:972–80.CrossRef
9.
Zurück zum Zitat Iwasa S, Nakajima TE, Nakamura K, et al. First-line fluorouracil-based chemotherapy for patients with severe peritoneal disseminated gastric cancer. Gastric Cancer. 2012;15:21–6.CrossRef Iwasa S, Nakajima TE, Nakamura K, et al. First-line fluorouracil-based chemotherapy for patients with severe peritoneal disseminated gastric cancer. Gastric Cancer. 2012;15:21–6.CrossRef
10.
Zurück zum Zitat Hara H, Kadowaki S, Asayama M, et al. First-line bolus 5-fluorouracil plus leucovorin for peritoneally disseminated gastric cancer with massive ascites or inadequate oral intake. Int J Clin Oncol. 2018;23:275–80.CrossRef Hara H, Kadowaki S, Asayama M, et al. First-line bolus 5-fluorouracil plus leucovorin for peritoneally disseminated gastric cancer with massive ascites or inadequate oral intake. Int J Clin Oncol. 2018;23:275–80.CrossRef
11.
Zurück zum Zitat Shitara K, Mizota A, Matsuo K, et al. Fluoropyrimidine plus cisplatin for patients with advanced or recurrent gastric cancer with peritoneal metastasis. Gastric Cancer. 2013;16:48–55.CrossRef Shitara K, Mizota A, Matsuo K, et al. Fluoropyrimidine plus cisplatin for patients with advanced or recurrent gastric cancer with peritoneal metastasis. Gastric Cancer. 2013;16:48–55.CrossRef
12.
Zurück zum Zitat Cunningham D, Starling N, Rao S, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36–46.CrossRef Cunningham D, Starling N, Rao S, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36–46.CrossRef
13.
Zurück zum Zitat Sawaki A, Yamaguchi K, Nabeya Y, et al. 5-FU/l-LV (RPMI) versus S-1 as first-line therapy in patients with advanced gastric cancer: a randomized phase III non-inferiority trial (ISO-5FU10 study group trial). Eur J Cancer Suppl. 2009;7:363.CrossRef Sawaki A, Yamaguchi K, Nabeya Y, et al. 5-FU/l-LV (RPMI) versus S-1 as first-line therapy in patients with advanced gastric cancer: a randomized phase III non-inferiority trial (ISO-5FU10 study group trial). Eur J Cancer Suppl. 2009;7:363.CrossRef
14.
Zurück zum Zitat Masuishi T, Kadowaki S, Kondo M, et al. FOLFOX as first-line therapy for gastric Cancer with severe peritoneal metastasis. Anticancer Res. 2017;37:7037–42.PubMed Masuishi T, Kadowaki S, Kondo M, et al. FOLFOX as first-line therapy for gastric Cancer with severe peritoneal metastasis. Anticancer Res. 2017;37:7037–42.PubMed
15.
Zurück zum Zitat Murad AM, Santiago FF, Petroianu A, Rocha PR, Rodrigues MA, Rausch M. Modified therapy with 5-fluorouracil, doxorubicin, and methotrexate in advanced gastric cancer. Cancer. 1993;72:37–41.CrossRef Murad AM, Santiago FF, Petroianu A, Rocha PR, Rodrigues MA, Rausch M. Modified therapy with 5-fluorouracil, doxorubicin, and methotrexate in advanced gastric cancer. Cancer. 1993;72:37–41.CrossRef
16.
Zurück zum Zitat Pyrhonen S, Kuitunen T, Nyandoto P, Kouri M. Randomised comparison of fluorouracil, epidoxorubicin and methotrexate (FEMTX) plus supportive care with supportive care alone in patients with non-resectable gastric cancer. Br J Cancer. 1995;71:587–91.CrossRef Pyrhonen S, Kuitunen T, Nyandoto P, Kouri M. Randomised comparison of fluorouracil, epidoxorubicin and methotrexate (FEMTX) plus supportive care with supportive care alone in patients with non-resectable gastric cancer. Br J Cancer. 1995;71:587–91.CrossRef
17.
Zurück zum Zitat Ishigami H, Fujiwara Y, Fukushima R, et al. Phase III trial comparing intraperitoneal and intravenous paclitaxel plus S-1 versus cisplatin plus S-1 in patients with gastric cancer with peritoneal metastasis: PHOENIX-GC trial. J Clin Oncol. 2018;36:1922–29.CrossRef Ishigami H, Fujiwara Y, Fukushima R, et al. Phase III trial comparing intraperitoneal and intravenous paclitaxel plus S-1 versus cisplatin plus S-1 in patients with gastric cancer with peritoneal metastasis: PHOENIX-GC trial. J Clin Oncol. 2018;36:1922–29.CrossRef
18.
Zurück zum Zitat Nishina T, Boku N, Gotoh M, et al. Randomized phase II study of second-line chemotherapy with the best available 5-fluorouracil regimen versus weekly administration of paclitaxel in far advanced gastric cancer with severe peritoneal metastases refractory to 5-fluorouracil-containing regimens (JCOG0407). Gastric Cancer. 2016;19:902–10.CrossRef Nishina T, Boku N, Gotoh M, et al. Randomized phase II study of second-line chemotherapy with the best available 5-fluorouracil regimen versus weekly administration of paclitaxel in far advanced gastric cancer with severe peritoneal metastases refractory to 5-fluorouracil-containing regimens (JCOG0407). Gastric Cancer. 2016;19:902–10.CrossRef
19.
Zurück zum Zitat Iwasa S, Goto M, Yasui H, et al. Multicenter feasibility study of combination therapy with fluorouracil, leucovorin and paclitaxel (FLTAX) for peritoneal disseminated gastric cancer with massive ascites or inadequate oral intake. Jpn J Clin Oncol. 2012;42:787–93.CrossRef Iwasa S, Goto M, Yasui H, et al. Multicenter feasibility study of combination therapy with fluorouracil, leucovorin and paclitaxel (FLTAX) for peritoneal disseminated gastric cancer with massive ascites or inadequate oral intake. Jpn J Clin Oncol. 2012;42:787–93.CrossRef
Metadaten
Titel
Fluoropyrimidine with or without platinum as first-line chemotherapy in patients with advanced gastric cancer and severe peritoneal metastasis: a multicenter retrospective study
verfasst von
Hiroyuki Arai
Satoru Iwasa
Narikazu Boku
Masahiro Kawahira
Hirofumi Yasui
Toshiki Masuishi
Kei Muro
Keiko Minashi
Shuichi Hironaka
Naoki Fukuda
Daisuke Takahari
Takako Eguchi Nakajima
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2019
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-019-5720-3

Weitere Artikel der Ausgabe 1/2019

BMC Cancer 1/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.