We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research ArticleOpen Accesscc iconby iconnc iconnd icon

Real-life use of ramucirumab in gastric cancer in Spain: the RAMIS study

    Federico Longo

    Hospital Universitario Ramón y Cajal, IRYCIS, CIBERONC, Madrid, 28034, Spain

    ,
    Mónica Jorge

    Hospital Álvaro Cunqueiro, Vigo (Pontevedra), 36213, Spain

    ,
    Ricardo Yaya

    Instituto Valenciano de Oncología (IVO), Valencia, 46009, Spain

    ,
    Ana Fernández Montes

    Complexo Universitario Hospitalario de Ourense, 32005, Spain

    ,
    Nieves Martínez Lago

    Complejo Hospitalario Universitario, A Coruña, 15006, Spain

    ,
    Elena Brozos

    Complejo Hospitalario Universitario de Santiago, Santiago de Compostela (La Coruña), 15706, Spain

    ,
    Jorge Aparicio

    Hospital Universitario y Politécnico La Fe, Valencia, 46026, Spain

    ,
    Guillermo Quintero

    Hospital Universitario Lucus Augusti, Lugo, 27003, Spain

    ,
    Eduardo Ceballos

    Hospital San Pedro de Alcántara, Cáceres, 10003, Spain

    ,
    Elvira Buxó

    Hospital Quirón Salud, Barcelona, 08023, Spain

    ,
    Ana Maria Lopez

    Hospital Universitario de Burgos, 09006, Spain

    ,
    Maria Luz Pellón

    Complexo Universitario Hospitalario de El Ferrol (La Coruña), 15405, Spain

    ,
    Raquel Molina

    Hospital Universitario Príncipe de Asturias, Alcalá de Henares (Madrid), 28805, Spain

    ,
    Laura Diaz-Paniagua

    Hospital Universitario de Getafe (Madrid), 28905, Spain

    ,
    Paula Cerdà

    Centro Médico Teknon, Barcelona, 08022, Spain

    ,
    Pedro Lopez Leiva

    Complejo Hospitalario de Jaén, 23007, Spain

    ,
    Alfonso Martín Carnicero

    Hospital de San Pedro, Logroño, 26006, Spain

    ,
    Antía Cousillas

    Complexo Universitario Hospitalario de Pontevedra, 36071, Spain

    ,
    Lorena Paris

    Centro Oncológico de Galicia, La Coruña, 15009, Spain

    ,
    Beatriz García-Paredes

    Hospital Clínico San Carlos, Madrid, 28040, Spain

    ,
    Carlos Romero

    Hospital POVISA, Vigo (Pontevedra), 36201, Spain

    ,
    María Ortega

    Department of Medical, Lilly, Madrid, 28108, Spain

    ,
    Alberto Molero

    Department of Medical, Lilly, Madrid, 28108, Spain

    ,
    Sergio de la Torre

    Department of Medical, Lilly, Madrid, 28108, Spain

    ,
    Min-Hua Jen

    European Statistics Group, Lilly, Surrey, GU206PH, UK

    &
    Silvia Díaz-Cerezo

    *Author for correspondence: Tel.: +34 663 320 665;

    E-mail Address: diaz_silvia@lilly.com

    Department of Medical, Lilly, Madrid, 28108, Spain

    Published Online:https://doi.org/10.2217/fon-2020-1216

    Abstract

    Aims: To obtain real-world data on ramucirumab use and effectiveness for the treatment of advanced gastric cancer (AGC) or gastroesophageal junction adenocarcinoma (GEJ). Methods: Observational, retrospective study carried out in 20 Spanish hospitals, in patients who started ramucirumab treatment between December 2015 and December 2018. Descriptive analysis was conducted for patient characteristics, treatment patterns and effectiveness outcomes. Results: Three hundred seventeen patients were included (93.7% treated with ramucirumab-paclitaxel and 6.3% with ramucirumab); age 62.5 (11.3) years; 66.9% male. Median progression-free survival and overall survival were 3.9 months (95% CI: 3.4–4.3) and 7.4 (95% CI: 6.4–8.9) in combination regimen and 2.0 (1.1–2.8) and 4.3 (95% CI: 1.9–7.3) in monotherapy, respectively. Conclusion: The study findings were consistent with available real-world studies and randomized clinical trials.

    Gastric cancer (GC) is the fifth most frequently diagnosed cancer and the third leading cause of cancer death [1]. Worldwide, it accounts for 5.7% [2] of all new cancer cases and 8.2% of all cancer-related deaths [1]. Although a gradual decline in its incidence has been globally registered in recent years, it remains a major health concern with 140,000 new cases diagnosed across Europe in 2012, resulting in 107,000 deaths [3]. In Spain, prevalence of gastric adenocarcinomas is 7810 cases per year, resulting in more than 5675 deaths per year [4]. Approximately 90% of GCs are adenocarcinomas that arise from the glands of the mucosa in the stomach [5]. The most common risk factors associated with GC include male sex (incidence is twice as high), advanced age, tobacco smoking, family history and possibly dietary habits, such as low consumption of fruits and vegetables or high intake of salty and smoked food [6,7].

    Surgery (subtotal or total gastrectomy) is the primary treatment option for GC, with recurrence common in up to 70% of patients [8]. Patients with inoperable locally advanced and/or metastatic (stage IV) disease receive systemic chemotherapy, the only treatment option that has shown a superior survival rate and improved quality of life (QoL) compared with best supportive care [7,9]. However, most patients relapse after first-line therapy. Literature suggests that 20–64% patients receive further treatment with second-line chemotherapy [8,10]. A second-line treatment is always justified in physically fit patients who are willing to receive treatment because it improves overall survival (OS), as well as symptom control and QoL [11].

    Ramucirumab (Cyramza®) is a second-line treatment option that selectively targets VEGF receptor (VEGFR)-2. It is a human IgG1 monoclonal antibody and the first biological agent for angiogenesis approved for GC by the US FDA and the EMA based on two randomized, double-blind, placebo-controlled Phase III trials (REGARD and RAINBOW) [12,13]. It demonstrated an ability to extend the life span and slow down tumor growth in patients with metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma progressing after first-line chemotherapy either as monotherapy or in combination with paclitaxel [12,13]. The REGARD trial compared ramucirumab with placebo and demonstrated improvement in OS (median OS of 5.2 vs 3.8 months), progression-free survival (PFS) (2.1 vs 1.3 months) and disease control rate (DCR) (49% vs 23%) [12]. The RAINBOW study demonstrated a longer OS (9.6 months vs 7.4 months) with ramucirumab plus paclitaxel compared to placebo plus paclitaxel along with improvement in PFS (4.4 months vs 2.9 months) and objective response rate (ORR) (28% vs 16%) [13].

    In Europe, available data outside the clinical trial setting come from the RAMoss study [14], which assessed the safety and effectiveness of ramucirumab in advanced GC (AGC) patients treated in Italy within a compassionate-use program before ramucirumab reimbursement authorization. Results from RAMoss study on OS (8 months), PFS (4.3 months) and ORR (20.2%) were consistent with those previously reported in the clinical trials, confirming the safety and efficacy of ramucirumab [14]. Outside of Europe, additional evidence comes from a real-world evidence study conducted in the USA [15] and from a study based on an expanded access program conducted in South Korea [16]; the authors also suggested in the conclusions that clinical outcomes observed in these settings were similar to data from Phase III clinical trials [12,13].

    Ramucirumab, as a single agent or combined with paclitaxel, has been marketed in Spain since December 2015 for the treatment of patients with AGC or GEJ adenocarcinoma with disease progression on or after prior fluoropyrimidine- and/or platinum-containing chemotherapy. The RAMIS observational study aimed to obtain real-world data on ramucirumab utilization in clinical practice in Spain as monotherapy or in combination with paclitaxel.

    Methods

    Study objectives

    The primary objectives of the study were to describe the clinical characteristics and the treatment patterns of the patients with AGC or GEJ adenocarcinoma treated with ramucirumab in clinical practice in Spain. The secondary objective aimed to describe the effectiveness of ramucirumab as a combination (with paclitaxel) or as monotherapy in current clinical practice. Further, the data were used to identify the predictive factors for effectiveness and assess effectiveness in specific subgroups of interest.

    Study design

    This was an observational, retrospective single-cohort study, based on medical chart review of patients with AGC or GEJ adenocarcinoma treated with ramucirumab in routine clinical practice. Twenty Spanish hospitals participated in the study.

    The observational study period was from December 2015 (date of ramucirumab commercialization in Spain) to the study initiation in each hospital (October–December 2018). Oncologist investigators or their collaborators collected the study variables of each patient from the time of ramucirumab treatment initiation until the index date (site study initiation) or death, whichever occurred first.

    The study was approved by the Ethics Committee of Hospital Ramón y Cajal in Madrid, Spain. It was conducted with the support of the Clinical Research Organization IQVIA for the following activities: protocol development, site monitoring and statistical analysis.

    Study population

    All patients with diagnosis of AGC or GEJ adenocarcinoma who had initiated treatment with ramucirumab since December 2015, aged ≥18 years and with complete medical record at the participating site or at least since the AGC or GEJ adenocarcinoma diagnosis, were included in the study. Patients who had participated in a clinical trial during the observational study were excluded.

    Study variables

    Demographic and clinical characteristics of patients included demographic variables, initial diagnosis of AGC or GEJ adenocarcinoma, tumor stage, diagnostic procedures, Eastern Cooperative Oncology Group (ECOG) performance status, histological and molecular characteristics of the tumor, comorbidities, history of previous treatments and laboratory variables. Descriptive data on treatment with ramucirumab were also collected, including the date of initiation of treatment with ramucirumab, regimen, dosage, number of cycles (as monotherapy or combination), regimen changes and dose adjustments.

    Effectiveness of ramucirumab was described in terms of median PFS and PFS rates at 3, 6, 9 and 12 months; median OS and OS survival rates at 3, 6 and 12 months; median time to progression (TTP); and tumor response for both measurable versus nonmeasurable disease (following RECIST v1.1 criteria) [17].

    For measurable tumors, responses were classified as complete response (CR), partial response (PR), progressive disease (PD) and stable disease (StD) (when an evaluation by imaging was available). In the case of nonmeasurable tumors, the response evaluation criteria used for classification were as follows: CR, noncomplete response/nonprogression and progression (as described in the medical record).

    Additionally, ORR, DCR, time to response and duration of response (DOR) were calculated for patients with measurable tumors evaluated for imaging.

    Statistical analysis

    A descriptive analysis of all included study variables was performed. Continuous variables were described as the number of patients with valid/missing observations, mean, standard deviation (SD), median, 25 and 75 percentiles (P25 and P75, respectively), minimum and maximum. Categorical variables were described by frequencies and related percentages.

    Descriptive analysis was also performed to describe the effectiveness variables in a set of subgroups of patients. Exploratory analysis of predictors of effectiveness was performed. Univariate Cox proportional hazard models among baseline characteristics and effectiveness variables were used to select the covariates to be introduced in the multivariate models. Multivariate Cox regression models were used to explore factors associated with clinical outcomes and was reported as hazard ratios (HR) with 95% CI and corresponding p-values. Exploratory analysis was performed to compare the results between different categories defined in each subgroup by parametric tests (Student's t-test) or nonparametric test (Mann–Whitney) for quantitative variables and X2-test for categorical variables. All analyses were performed using Kaplan–Meier (KM) survival analysis and the plots of survival curve were produced. DCR was calculated by adding the CR, PR and StD, as the best response during the study period. The method of KM was used to estimate the OS and PFS of patients stratified by therapy at 3, 6, 9 and 12 months. All effectiveness variables were analyzed separately for patients treated with monotherapy and combination therapy. Statistical analysis was performed using SAS version 9.4.

    Results

    Study population

    A total of 318 patients treated with ramucirumab beginning in December 2015 were included in the database. One patient was excluded from final analysis because the treatment was initiated after the start of the study data collection; thus, the final number of patients eligible for full analysis set (FAS) were 317. Among the FAS, 297 (93.7%) initiated ramucirumab as combination regimen (first cycle of ramucirumab received in combination with paclitaxel), and 20 (6.3%) were prescribed monotherapy regimen (first cycle of ramucirumab received as monotherapy).

    Patient & clinical characteristics

    Patient characteristics

    The mean (SD) age of enrolled patients was 62.5 (11.3) years and majority of the patients were males (66.9%) with mean (SD) BMI of 25.4 (29.4) kg/m2 at ramucirumab treatment initiation. Clinically relevant chronic comorbidities at ramucirumab treatment initiation were present in 62.5% of patients. The most common comorbidities were hypertension (29.3%), dyslipidemia (23.0%), diabetes mellitus (12.0%) and other cardiovascular diseases (9.5%) (Table 1).

    Table 1. Sociodemographic and clinical characteristics of enrolled patients.
    Analysis variableCombination regimen (n = 297)Monotherapy regimen (n = 20)Total (n = 317)
    Gender
    Male, n (%)204 (68.7%)8 (40.0%)212 (66.9%)
    Female, n (%)93 (31.3%)12 (60.0%)105 (33.1%)
    Age (year) at treatment start with ramucirumab, mean (SD)62.00 (11.0)69.30 (13.3)62.46 (11.3)
    BMI (kg/m2) at treatment start with ramucirumab, mean (SD)25.56 (30.2)23.53 (4.9)25.44 (29.4)
    Percentage of weight loss (from initiation of first line to initiation of ramucirumab), mean (SD)4.17 (9.1)7.72 (13.9)4.36 (9.5)
    Smoking status
    Current smoker, n (%)51 (17.2%)3 (15.0%)54 (17.0%)
    Ex-smoker, n (%)108 (36.4%)2 (10.0%)110 (34.7%)
    Nonsmoker, n (%)113 (38.0%)15 (75.0%)128 (40.4%)
    Unknown or not available, n (%)25 (8.4%)25 (7.9%)
    Presence of comorbidities
    No, n (%)113 (38.0%)6 (30.0%)119 (37.5%)
    Yes, n (%)184 (62.0%)14 (70.0%)198 (62.5%)
    Comorbidities
    Total, n (%)297 (100%)20 (100%)317 (100%)
    Hypertension82 (27.6%)11 (55.0%)93 (29.3%)
    Dyslipidemia65 (21.9%)8 (40.0%)73 (23.0%)
    Diabetes mellitus34 (11.4%)4 (20.0%)38 (12.0%)
    Depression16 (5.4%)2 (10.0%)18 (5.7%)
    Other GI disease15 (5.1%)2 (10.0%)17 (5.4%)
    Cardiovascular disease26 (8.8%)4 (20.0%)30 (9.5%)
    Respiratory disease19 (6.4%)2 (10.0%)21 (6.6%)
    Other malignancy (including solid tumors, leukemia or lymphoma)17 (5.7%)1 (5.0%)18 (5.7%)
    Other comorbidities37 (12.5%)3 (15.0%)40 (12.6%)
    ECOG PS
    ECOG 0, n (%)70 (23.6%)1 (5.0%)71 (22.4%)
    ECOG 1§, n (%)189 (63.6%)11 (55.0%)200 (63.1%)
    ECOG 2, n (%)28 (9.4%)5 (25.0%)33 (10.4%)
    ECOG 3#, n (%)1 (0.3%)1 (5.0%)2 (0.6%)
    Unknown or not available, n (%)9 (3.0%)2 (10.0%)11 (3.5%)
    TNM staging
    T399 (33.8%)4 (20.0%)103 (32.9%)
    T488 (30.0%)4 (20.0%)92 (29.4%)
    N259 (20.1%) 59 (18.8%)
    N356 (19.1%)3 (15.0%)59 (18.8%)
    M1181 (61.4%)15 (75.0%)196 (62.2%)
    Metastatic/locally advanced unresectable disease detection
    At diagnosis, n (%)193 (65.2%)18 (90.0%)211 (66.8%)
    After radical surgery, n (%)103 (34.8%)2 (10.0%)105 (33.2%)
    Metastatic sites at treatment initiation (n)
    0, n (%)12 (4.0%)1 (5.0%)13 (4.1%)
    1, n (%)133 (44.8%)10 (50.0%)143 (45.1%)
    2, n (%)96 (32.3%)7 (35.0%)103 (32.5%)
    ≥3, n (%)56 (18.9%)2 (10.0%)58 (18.3%)
    Location of metastatic sites (multiresponse)
    Peritoneal, n (%)161 (54.2%)7 (35.0%)168 (53.0%)
    Liver, n (%)98 (33.0%)11 (55.0%)109 (34.4%)
    Lung, n (%)53 (17.8%)4 (20.0%)57 (18.0%)
    Adenopathies, n (%)47 (15.8%)2 (10.0%)49 (15.5%)
    Lymphatic, n (%)39 (13.1%)2 (10.0%)41 (12.9%)
    Bone, n (%)32 (10.8%)1 (5.0%)33 (10.4%)
    Other locations, n (%)51 (17.2%)2 (10.0%)53 (16.7%)
    HER2 status
    Positive, n (%)43 (14.5%)2 (10.0%)45 (14.2%)
    Negative, n (%)237 (79.8%)16 (80.0%)253 (79.8%)
    Not available, n (%)17 (5.7%)2 (10.0%)19 (6.0%)
    Presence of ascites
    Yes, n (%)84 (28.3%)7 (35.0%)91 (28.7%)
    No, n (%)203 (68.4%)13 (65.0%)216 (68.1%)
    Not available, n (%)10 (3.4%) 10 (3.2%)
    Measurability
    Measurable, n (%)224 (77.5%)15 (83.3%)239 (77.9%)
    Nonmeasurable, n (%)65 (22.5%)3 (16.7%)68 (22.1%)
    Time since initial diagnosis of AGC or GEJ to first ramucirumab dose (months)34.4 (24.4)33.1 (29.4)34.3 (24.7)
    Time since first diagnosis of metastatic/locally advanced unresectable disease to start ramucirumab (months)12.4 (16.4)14.7 (15.2)12.5 (16.3)

    Includes comorbidities presented in <5% of patients in the total sample (anemia, osteoporosis, hepatic disease, renal disease, cerebrovascular disease, thromboembolic phenomena, cognition impairment/dementia, infectious disorder) as well as information collected directly as ‘others’.

    Fully active, able to carry on all predisease performance without restriction.

    §Restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature, e.g., light housework, office work.

    Ambulatory and capable of all self-care but unable to carry out any work activities; up and active for >50% of waking hours.

    #Capable of only limited self-care; confined to bed or chair more than 50% of waking hours.

    AGC: Advanced gastric cancer; ECOG: Eastern Cooperative Oncology Group; GEJ: Gastroesophageal junction adenocarcinoma; GI: Gastrointestinal; SD: Standard deviation.

    Tumor characteristics

    The majority of the patients had gastric adenocarcinoma (77.6%); primary tumor was still present in 64.0% of the patients. Tumor grade was poorly (43.2%), moderately (24.0%) or well differentiated (6.9%). Histological subtype distribution was intestinal (41.0%), diffuse (30.3%) and mixed (3.8%). TNM (tumor node metastasis) staging revealed a high percentage of patients with T3 (32.9%) and T4 (29.4%), N2 (18.8%) and N3 (18.8%) and M1 (62.2%) disease. A majority of the patients had measurable tumor (77.9%) (Table 1).

    Diagnostic characteristics

    The most commonly used diagnostic procedures were computerized tomography (CT) (97.2%) and endoscopy (94.6%).

    The mean time since initial diagnosis of AGC or GEJ adenocarcinoma to start of ramucirumab treatment was 18.6 (SD: 22.2) months. Metastatic/locally advanced unresectable disease was detected at initial diagnosis in 66.8% patients and occurred after radical surgery in 33.2%. More than three-quarters of the patients had one or two metastatic sites (77.6%) at the start of the treatment with ramucirumab. A total of 14.2% of patients with AGC/GEJ adenocarcinoma were HER2 positive, and ascites were present in 28.7% of patients (Table 1).

    ECOG Performance Status

    Patients included in the study were assessed as ECOG Performance Status 0–3 [18]. Higher proportion of patients in both groups (combination and monotherapy) belonged to ECOG 1 category (63.6% and 55.0%, respectively). Patients in monotherapy group had a higher proportion of patients in the ECOG categories 2 and 3 compared with the combination group (Table 1).

    Laboratory values

    Various laboratory parameters were analyzed at treatment initiation. The mean (SD) neutrophil-to-lymphocyte ratio was 4.5 (14.02). The mean (SD) hemoglobin level was 11.7 (1.6) g/dl and albumin level was 3.7 (0.6) g/dl; 30.0% of patients had values <3.5 g/dl. The mean (SD) values for C-reactive protein was 0.5 (0.6) mg/dl and 56.3% showed levels >0.2 mg/dl. The mean (SD) level of carcinoembryonic antigen (CEA) was 64.0 (287.4) ng/ml (Table 2).

    Table 2. Laboratory values at treatment initiation.
    Analysis variableCombination regimen (n = 297)Monotherapy regimen (n = 20)Total (N = 317)
    Neutrophils (per mcl)   
    Mean (SD)4359 (2500)6412 (7589)4477 (3040)
    Med (P25; P75)3700 (2700; 5400)4303 (3177; 5300)3790 (2800; 5390)
    Lymphocytes (per mcl)   
    Mean (SD)1632 (885)1429 (543)1621 (869)
    Med (P25; P75)1575 (1090; 2000)1510 (1000; 1700)1570 (1090; 2000)
    NLR (calculated)   
    Mean (SD)4.4 (14.1)6.7 (13.4)4.5 (14.0)
    Med (P25; P75)2.5 (1.7; 3.9)3.5 (2.2; 5.0)2.5 (1.7; 4.0)
    Haemoglobin (g/dl)   
    Mean (SD)11.7 (1.6)11.1 (1.4)11.7 (1.6)
    Med (P25; P75)12.0 (11.0; 13.0)11.0 (10.0; 12.0)12.0 (11.0; 13.0)
    Albumin level (g/dl)   
    Mean (SD)3.8 (0.6)3.6 (0.5)3.7 (0.6)
    Med (P25; P75)3.8 (3.3; 4.2)3.6 (3.2; 4.0)3.8 (3.3; 4.2)
    CRP (mg/dl)   
    Mean (SD)0.50 (0.65)0.45 (0.1)0.5 (0.6)
    Med (P25; P75)0.3 (0.1; 0.6)0.5 (0.4; 0.5)0.4 (0.1; 0.6)
    LDH (U/L)   
    Mean (SD)380.3 (324.8)427.4 (229.2)383.4 (319.2)
    Med (P25; P75)291.0 (200.0; 419.0)381.5 (253.5; 635.5)305.0 (200.0; 432.0)
    Platelet count (per mcl)   
    Mean (SD)2,226,067 (98,476)223,765 (82,361)222,673 (97,505)
    Med (P25; P75)209,500 (154,000; 274,000)204,000 (172,000; 265,000)209,000 (156,000; 273,000)
    CEA (ng/ml)   
    Mean (SD)66.6 (294.9)16.8 (22.6)64 (287.4)
    Med (P25; P75)4.1 (1.9; 16.2)7.4 (0.9; 20.7)4.3 (1.9; 17.3)

    CEA: Carcinoembryonic antigen; Med: Median; NLR: Neutrophil to lymphocyte ratio; SD: Standard deviation.

    Treatment pattern

    Treatment history

    A total of 97.2% patients received chemotherapy, 47.3% underwent surgery, 13.9% received radiotherapy and 10.4% received targeted therapy before ramucirumab initiation. Chemotherapy was administered as first line in 75.6% patients (73.4% and 26.6% patients received doublet and triplet regimen, respectively), neoadjuvant in 28.9% (30.4% and 69.6% patients received doublet and triplet regimen, respectively) and adjuvant in 21.4% patients (61.0% and 39.0% patients received doublet and triplet regimen, respectively). Out of the 26.6% of patients who received triplet chemotherapy in first line, 13.3% included taxanes. Mean duration of taxanes treatment in those patients was 147 (126.7) days (Table 3).

    Table 3. History of previous treatments received for advanced gastric cancer or gastroesophageal junction adenocarcinoma.
    Analysis variableCombination regimen (n = 297)Monotherapy regimen (n = 20)Total (N = 317)
    Type of surgery   
    Total, n (%)145 (48.8%)5 (25.0%)150 (47.3%)
    Gastrectomy, n (%)121 (83.4%)4 (80.0%)125 (83.3%)
    Palliative, n (%)24 (16.6%)1 (20.0%)25 (16.7%)
    Radiotherapy, total, n (%)41 (13.8%)3 (15.0%)44 (13.9%)
    Type of chemotherapy   
    Total, n (%)288 (97.0%)20 (100%)308 (97.2%)
    Neoadjuvant, n (%)85 (29.5%)4 (20.0%)89 (28.9%)
    – Doublets, n (%)22 (28.6%)2 (100%)24 (30.4%)
    – Triplets, n (%)55 (71.4%) 55 (69.6%)
    Adjuvant, n (%)64 (22.2%)2 (10.0%)66 (21.4%)
    – Doublets, n (%)36 (62.1%) 36 (61.0%)
    – Triplets, n (%)22 (37.9%)1 (100%)23 (39.0%)
    First line, n (%)216 (75.0%)17 (85.0%)233 (75.6%)
    – Doublets, n (%)141 (72.7%)11 (84.6%)152 (73.4%)
    – Triplets, n (%)53 (27.3%)2 (15.4%)55 (26.6%)
    Disease progression after treatment initiation (first line), n (%)216 (100%)17 (100%)233 (100%)
    ≤6 months119 (55.1%)8 (47.1%)127 (54.5%)
    ≥6 months97 (44.9%)9 (52.9%)106 (45.5%)
    Taxane use, yes37 (12.8%)4 (20.0%)41 (13.3%)
    Taxane duration (days)   
    Mean (standard deviation)146.46 (131.60)154.00 (1.41)147.00 (126.65)
    Median (P25; P75)105.0 (50.0; 188.0)154.0 (153.0; 155.0)107.0 (64.0; 186.5)
    Targeted therapy, total, n (%)32 (10.8%)1 (5.0%)33 (10.4%)

    Treatment with ramucirumab

    Most (93.7%) patients initiated ramucirumab treatment in combination; 20 (6.3%) in monotherapy. The median duration of ramucirumab treatment according KM estimator was 3.2 months (95% CI: 2.8–3.4 months). The median number of cycles (28 days) received was 2.5 for patients initiating monotherapy and 4.0 for combination therapy.

    According to ramucirumab treatment, 297 (93.7%) patients received combination therapy (always with paclitaxel); 265 (83.6%) of total patients received it during all treatment duration (median of three cycles); 32 (10.1%) received combination followed by monotherapy (three cycles in combination and three cycles in monotherapy) with a mean (SD) time of 4.1 (3.7) months in combination before changing to monotherapy. Nineteen patients (6.0%) received ramucirumab monotherapy (median of three cycles). One patient received monotherapy and then combination therapy. A total of seven patients (2.2%), all treated with a combination regimen, needed dose adjustment of ramucirumab, and the mean (SD) time to first dose adjustment was 5.5 months (3.4 months).

    The dose of ramucirumab combination and monotherapy was 8 mg/kg on days 1 and 15 of a 28-day cycle during all treatment in 98.5% and 100% patients, respectively.

    Effectiveness with ramucirumab treatment

    Overall survival

    The median OS was 7.2 months (95% CI: 6.2–8.5 months) for overall population. OS rate at 3 months was 83.1%, 58.7% at 6 months and 33.4% at 12 months (Figure 1). The median OS for patients treated with ramucirumab in a combination regimen was 7.4 months (95% CI: 6.4–8.9 months) and 4.3 months (95% CI: 1.9–7.3 months) for patients with a monotherapy regimen.

    Figure 1. Kaplan–Meier estimates of survival function (overall survival) for both combination therapy and monotherapy.

    Progression-free survival

    The median PFS was 3.8 months for overall population (3.9 months for combination therapy and 2.0 months for monotherapy group) (Table 4). PFS rate at 3 months was 58.8%, 29.7% at 6 months and 11.1% at 12 months (Figure 2). PFS rates for combination and monotherapy, respectively, were 60.9% and 26.3% at 3 months, 30.6% and 15.8% at 6 months and 11.6% and 0% at 12 months.

    Table 4. Overall survival and progression-free survival according to sociodemographic and clinical characteristics of patients (total sample size: n = 317).
    CovariateMedian OSMedian PFS
    Overall, median7.2 (6.2–8.5)3.8 (3.3–4.2)
    Time to progressive disease on first-line therapy (chemotherapy and targeted therapy)
    ≤6 months5.7 (4.5–7.4)3.0 (2.7–3.8)
    >6 months7.7 (6.1–11.1)4.3 ([3.3–5.4)
    Disease measurement  
    Measurable disease8.9 (7.0–10.3)4.1 (3.6–4.7)
    Nonmeasurable disease5.0 (4.0–6.4)2.8 (2.3–3.6)
    Gender  
    Female6.4 (5.2–9.6)3.9 (2.9–4.9)
    Male7.4 (6.3–9.0)3.7 (3.0–4.3)
    Age range (years)  
    <656.6 (5.6–7.5)3.8 (3.0–4.5)
    ≥658.5 (6.4–10.5)3.8 (2.9–4.3)
    ECOG PS status  
    ECOG 010.2 (8.5–20.9)5.5 (4.9–6.3)
    ECOG 16.8 (5.7–7.9)3.5 (2.9–3.9)
    ECOG ≥24.0 (2.0–5.2)2.5 (1.9–3.9)
    Unknown or not available5.4 (1.9–NA)2.1 (1.0–15.0)
    Previous weight loss  
    No weight loss6.7 (4.5–14.7)3.5 (2.8–4.1)
    Weight loss <10%8.3 (6.4–9.7)4.1 (3.4–4.9)
    Weight loss ≥10%7.8 (5.2–12.1)3.7 (2.3–4.5)
    Tumour site  
    Gastric adenocarcinoma6.8 (6.1–8.5)3.8 (3.1–4.3)
    GEJ adenocarcinoma7.5 (5.5–12.2)3.7 (2.7–4.9)
    Previous first-line chemotherapy  
    Doublets7.5 (6.4–9.7)3.8 (3.4–4.5)
    Triplets7.2 (5.5–9.6)3.4 (2.9–4.5)
    Histological subtype  
    Diffuse6.0 (4.4–7.2)3.4 (2.8–4.3)
    Intestinal9.2 (6.7–11.9)3.7 (3.0–4.5)
    Mixed13.3 (1.9–NA)7.3 (1.6–13.3)
    Unknown or not available7.4 (5.4–10.2)3.8 (2.9–4.8)
    Number of metastatic sites  
    Metastatic sites ≤2, (n)7.8 (6.2–9.7)3.9 (3.4–4.3)
    Metastatic sites ≥3, (n)6.4 (4.5–7.3)3.4 (2.6–4.2)
    Peritoneal metastases  
    No9.6 (7.3–12.4)3.9 (3.4–5.0)
    Yes6.1 (5.2–6.8)3.5 (2.9–4.1)
    Previous gastrectomy  
    No6.6 (5.5–7.8)3.5 (2.9–4.0)
    Yes9.2 (6.3–12.8)4.3 (3.5–5.0)
    Presence of thromboembolic phenomena  
    No7.0 (6.1–8.4)3.8 (3.2–4.2)
    YesNA3.6 (1.8–9.6)
    Status at diagnosis  
    Metastatic (M1)6.4 (5.3–7.7)3.3 (2.9–3.9)
    Locally advanced unresectable disease (T3–4 or N1–3)9.5 (6.8–13.3)5.0 (3.8–6.1)
    Other status7.4 (4.0–18.4)3.1 (1.7–7.8)
    Metastatic/locally advanced unresectable disease detection  
    After radical surgery8.5 (6.4–12.2)4.5 (3.7–5.7)
    At diagnosis6.6 (5.5–7.8)3.4 (2.9–4.0)
    HER2 status  
    Negative6.7 (6.1–8.3)3.7 (3.0–4.1)
    Not available5.5 (3.2–7.8)2.8 (2.3–4.3)
    Positive9.7 (7.4–22.7)4.9 (3.5–7.4)
    Previous treatment with taxane  
    No7.0 (6.1–8.4)3.8 (3.4–4.2)
    Yes8.5 (5.1–9.5)2.9 (2.5–4.9)
    Tumor grade  
    Moderately differentiated7.4 (6.1–11.0)3.7 (2.9–5.4)
    Poorly differentiated6.7 (5.5–8.8)3.8 (2.9–4.5)
    Unknown or missing6.6 (4.4–8.5)3.4 (2.9–4.1)
    Well differentiated13.0 (4.0–24.9)4.0 (2.7–7.4)
    Presence of ascites  
    No9.5 (7.7–11.9)4.1 (3.5–4.9)
    Not available3.1 (0.5–6.8)2.3 (0.5–4.3)
    Yes5.1 (4.3–6.0)3.0 (2.7–3.9)
    Helicobacter pylori  
    No6.7 (5.5–9.5)3.1 (2.7–3.9)
    Not available7.5 (6.0–9.7)4.3 (3.4–4.9)
    Yes6.7 (5.2–9.4)3.9 (3.5–5.7)
    Smoking status  
    Current smoker6.8 (4.9–11.0)3.9 (3.0–5.4)
    Ex-smoker8.5 (7.0–12.8)4.5 (3.2–5.2)
    Nonsmoker6.4 (5.2–8.9)3.5 (2.8–4.1)
    Unknown or not available5.5 (4.0–8.2)2.8 (1.7–4.0)
    Months with ramucirumab  
    Less than 1 month2.6 (1.8–4.3)1.2 (1.0–1.6)
    Between 1 and 3 months4.1 (3.6–5.4)2.5 (2.3–2.6)
    Between 3 and 5 months6.4 (5.5–8.3)4.1 (3.9–4.5)
    More than 5 months20.6 (13.0–22.7)8.5 (7.3–10.4)
    Ramucirumab in combination, ECOG PS-0/1 and effectiveness variables assessed by imaging10.3 (8.5–12.3)4.9 (3.9–5.4)

    ECOG: Eastern Cooperative Oncology Group; GEJ: Gastroesophageal junction; NA: Not available; OS: Overall survival; PFS: Progression-free survival; PS: Performance status.

    Figure 2. Kaplan–Meier estimates of progression-free survival function for both combination and monotherapy.

    Time to progression

    Median TTP was 4.3 months for overall population (95% CI: 3.7–4.9 months) with 4.5 months (95% CI: 3.8–5.0) for combination regimen and 2.5 months (95% CI: 1.2–4.3 months) for monotherapy.

    Objective tumor response

    For patients with measurable disease (n = 239), CR and PR was observed in 2.9% (n = 7) and 17.6% (n = 42) of the patients, respectively (ORR: 19.2%). For nonmeasurable disease (n = 68), no patients obtained a CR, but 35.3% of patients obtained a best overall response of noncomplete response/nonprogression.

    Time to response

    Response was achieved by 46 patients for whom mean (SD) time to first response was 2.95 (2.37) month or 2.5 months as median.

    Duration of response

    Forty-six patients obtained response (CR/PR). During follow-up period, a total of 28 patients (60.9%) progressed after PR/CR response. The median time for DOR (since response to first progression) was 5.6 months (95% CI: 3.1–7.9 months). The probability of being progression free after response was 75.7% at 3 months decreasing to 44.7% and 20.6% at 6 and 12 months, respectively.

    Subgroup analysis

    Table 4 describes KM survival analysis of PFS and OS according to different sociodemographic and clinical characteristics. Median time of OS was longer for patients with ECOG 0 compared with ECOG ≥2 (10.2 [95% CI: 8.5–20.9] vs 4.0 [95% CI: 2.0–5.2] months) and for patients with measurable versus nonmeasurable disease (8.9 [95% CI: 7.0–10.3] vs 5.0 [95% CI: 4.0–6.4] months). Median time of OS was shorter for patients with peritoneal metastatic sites versus no such sites (6.1 [95% CI: 5.2–6.8] vs 9.6 [95% CI: 7.3–12.4]) and for patients with presence of ascites versus no presence (5.7 [95% CI: 4.3–6.0] vs 9.5 [95% CI: 7.7–11.9]). The median OS in patients treated with ramucirumab in combination, ECOG PS-0/1 and effectiveness variables assessed by imaging was 10.3 months (95% CI: 8.5–12.3 months).

    Median time of PFS in patients with ECOG 0 was 5.55 (95% CI: 4.9–6.3 months), with measurable disease was 4.1 (95% CI: 3.6–4.7) and in patients without presence of ascites was 4.1 (95% CI: 3.5–4.9). The median PFS according to patients with tumor response evaluated by imaging, treated with combination regimen and ECOG 0/1 was 4.9 months (95% CI: 3.9–5.4 months).

    Predictor of response

    According to multivariable Cox proportional hazards regression model, higher hazard of death was related to nonmeasurable disease compared with measurable disease (HR: 1.7 [1.2–2.3; 95% CI]), ECOG 1 or ECOG ≥2 status compared with ECOG 0 performance status (HR: 1.7 and 2.6, respectively), and patients with ascites compared with patients without ascites (HR: 1.5; 95% CI: 1.1 to 2.1). Higher hazard of progression (or death) was related to monotherapy versus combination therapy (HR: 2.1; 95% CI: 1.2–3.4), nonmeasurable versus measurable disease (HR: 1.8 [95% CI: 1.4–2.4]), ECOG 1 or ECOG ≥2 versus to ECOG 0 (HR: 1.6; 95% CI: 1.2–2.2 and HR: 2.3; 95% CI: 1.5–3.7) and patients with 3 or more versus ≤2 metastatic sites (HR: 1.5; 95% CI: 1.1–2.0). Additional information is included in Supplementary Tables 1 & 2).

    Discussion

    Real-world evidence about the treatment patterns and effectiveness of drugs is increasingly being requested by regulatory bodies, payers and healthcare professionals to obtain additional data beyond predefined criteria in clinical trials.

    Several observational studies have confirmed ramucirumab's safety and effectiveness in real life, with survival data similar to that obtained in Phase III trials [14–16]. However, no observational studies had been performed in Spain. The present study is the largest observational study with ramucirumab in gastric cancer conducted in Europe and the second worldwide, after the North American study [15], in terms of number of patients. As occurs with other real-life studies, its broader inclusion criteria with respect to Phase III trials [12,13] allow the generation of evidence in patients not represented or underrepresented in the latter, such as patients with comorbidities. Although some baseline characteristics in the RAMIS study were similar to patients participating in clinical trials regarding mean values of age, gender, AGC diagnosis with primary tumor still present and percentage of patients with ECOG 1, in general, the patient population in the present study had a worse prognosis, with a higher presence of proven risk factors such as ECOG 2/3, peritoneal metastases, clinically relevant chronic comorbidities (e.g., hypertension, dyslipidemia), weight loss >10% and nonmeasurable disease [19].

    In first line, 73% of patients had been treated with doublet (without taxanes) and 27% with triplet chemotherapy (i.e., taxanes in combination with a platinum and 5-fluorouracil-based regimen). Triplet chemotherapy with taxanes can be a good treatment option for patients with high tumor burden and strong pressure to achieve remission, but they should only be administered in select patients with good general health because this modality lacks survival benefit and confers high toxicity [20]. On the other hand, clinical practice seems to reflect physicians' perception that a small number of patients with GC may benefit clinically from reexposure to taxanes, similar to other cancer treatment approaches (i.e., hormone-sensitive breast cancer or platinum-sensitive ovarian cancer). The RAMIS study suggests that ramucirumab outcomes after a doublet chemotherapy were similar to those observed after triplet chemotherapy, in both median PFS (3.8 vs 3.4 months) and OS (7.5 vs 7.2 months), thus being an appropriate choice in either setting.

    After first-line chemotherapy, 93.7% patients started ramucirumab treatment in combination with paclitaxel, and 89.2% of these remained in combination during all treatment period. Only 2.2% of patients needed a ramucirumab dose adjustment, which was comparable to dose adjustments (5%) needed in the RAINBOW trial [13], suggesting good tolerability of ramucirumab in combination therapy in real-life conditions.

    Although cross-trial comparisons should be made with caution, it seems that ramucirumab effectiveness results in both median time and rates were in line with previous trials [12,13], finding better outcomes with a combination regimen versus monotherapy (median PFS 3.9 months [95% CI: 3.4–4.3 months] vs 2.0 months [95% CI: 1.1–2.8 months] and median OS 7.4 months [95% CI: 6.4–8.9 months] vs 4.3 months [95% CI: 6.4–8.9 months]).

    Similarly, ramucirumab treatment in combination with paclitaxel showed 4.4 months for PFS and 9.6 months for OS in the RAINBOW trial [13] and 2.1 months for PFS and 5.2 months for OS in REGARD trial [12]. Other treatments such as irinotecan or docetaxel have shown a median OS of 4.0 months in the AIO20 and of 5.2 months in the COUGAR-02 clinical trials, respectively [21,22].

    All observed responses in our study were in the combination group; results are also in line with clinical trials (3% of patients obtained a PR/CR in the REGARD trial and a 28% response was observed in the RAINBOW trial) [12,13]. ORR values in RAMIS (19.2%) were comparable to the similar observational study RAMoss (20.2%) and slightly lower than in RAINBOW (28%). The difference with RAINBOW could be related to including ECOG PS 2–3 patients.

    The patients more similar to those included in RAINBOW trial (173 patients with ECOG PS 0/1, treated with ramucirumab combined with paclitaxel and effectiveness assessed by imaging) showed better results than the total sample, with median PFS and OS values of 4.9 and 10.3 months, respectively.

    Predictive factors for effectiveness, consistent with previous studies, were ramucirumab in combination regimen, ECOG PS 0, few metastatic sites and absence of ascites [12–16]. Presence of measurable disease was found to be a positive factor, unlike previous studies [12–14]. HER2+ had not been evaluated in multivariate analysis in previous studies [12–16] and in RAMIS was found to be a predictor for survival (HER2+ versus test not available).

    The RAMIS study allowed evaluation of some subgroups of patients on which there is limited information in the literature, such as HER2+ patients. There are published data on a small cohort study of ten HER2+ patients who responded well and for long periods of time when receiving ramucirumab-based therapy in the second-line setting. The authors argued that this observation was consistent with an interaction between angiogenesis and HER2 signaling and that trastuzumab resistance might be reversed with the inhibition of the angiogenesis pathway [23].

    Results in the RAMIS and RAMoss studies were consistent in this subpopulation, with median PFS were 4.9 versus 4.4 months, respectively, and median OS were 9.7 versus 7.9 months, respectively.

    The present study has several limitations, mainly those related to its retrospective design, such as the possible lack of information registered in the medical charts and the fact that the effectiveness variables could not be complemented with quality-of-life data or other patient-reported outcomes not systematically collected in clinical practice. Nevertheless, this design guarantees that the data followed the usual clinical practice, without additional intervention. On the other hand, no safety information was collected; however, data coming from an ongoing ramucirumab post-authorization safety and effectiveness study will help physicians reveal this important information [24].

    Additionally, in RAMIS study, 13.9% of patients were still receiving treatment at the time of data collection, so final figures could vary slightly. Finally, the effectiveness results presented by subgroups must be interpreted with caution because no formal sample size calculation was done to make any comparison, including the analysis between ramucirumab in monotherapy versus ramucirumab plus paclitaxel. In addition, it must be taken into consideration that the rest of the subgroup analysis included both RAM-treated and RAM + PTX-treated patients, with the potential impact on the results of the distribution of patients by treatment schedule.

    In conclusion, ramucirumab use and effectiveness are consistent with available real-world studies and randomized clinical trials, given the presence of poor outcome indicators in the studied population.

    Summary points
    • Until now, there have been limited published data on the use of ramucirumab in clinical practice. The present study (RAMIS) constitutes the largest observational study with ramucirumab in gastric cancer conducted in Europe, with the inclusion of 317 patients from 20 Spanish hospitals.

    • The patients' baseline characteristics were as follows: 66.9% male; mean (standard deviation [SD]) age 62.5 (11.3) years; 77.6% AGC; mean (SD) time since metastatic disease 1.0 (1.4) year; ECOG status 0 (22.4%), 1 (63.1%) and ≥2 (11%); and 77.9% measurable disease.

    • Ramucirumab treatment initiation occurred 18.6 months after the diagnosis, mostly in patients with metastatic diseases (77.6%), previously treated with chemotherapy, surgery, radiotherapy or targeted therapy (97.2%, 47.3%, 13.9% and 10.4%, respectively).

    • Ramucirumab was administered in combination with paclitaxel in 93.7% of patients (297/317) and in monotherapy in 6.3% (20/217). Main effectiveness outcomes in these two groups were as follows: median PFS and OS 3.9 months (95% CI: 3.4–4.3) and 7.4 (95% CI: 6.4–8.9) in combination regimen and 2.0 (1.1–2.8) and 4.3 (95% CI: 1.9–7.3) in monotherapy, respectively.

    • For patients with measurable disease (n = 239), CR and PR was observed in 2.9% (n = 7) and 17.6% (n = 42) of the patients, respectively (ORR: 19.2%). For nonmeasurable disease (n = 68), no patients obtained a CR, but 35.3% of patients obtained a best overall response of noncomplete response/nonprogression.

    • Multivariate Cox proportional hazards regression models showed that higher hazard of progression was related to monotherapy versus combination therapy (HR: 2.1; 95% CI: 1.2–3.4), nonmeasurable versus measurable disease (HR: 1.8; 95% CI: 1.4–2.4), ECOG 1 versus ECOG 0 (HR: 1.6; 95% CI: 1.2–2.2), ECOG 2 versus ECOG 0 (HR: 2.3; 95% CI: 1.5–3.7) and ≥3 versus ≤2 metastatic sites (HR: 1.5; 95% CI: 1.1–2.0).

    • In conclusion, the population included in this study had a worse prognosis with respect to the patients in the clinical trials and the RAMoss study ( patients treated with ramucirumab in a compassionate use program) due to the higher presence of proven risk factors such as ECOG 2/3, peritoneal metastases, clinically relevant chronic comorbidities (e.g., hypertension, dyslipidemia) and nonmeasurable disease. Treatment patterns and ramucirumab effectiveness data were generally consistent with results of clinical trials and previous studies in real life conditions.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.futuremedicine.com/doi/suppl/10.2217/fon-2020-1216

    Author contributions

    All authors meet the International Committee of Medical Journal Editors criteria for authorship for this article, take responsibility for the integrity of the work as a whole and have given their approval for this version to be published.

    Acknowledgments

    The authors thank all study participants: patients, investigators and institutions.

    Financial & competing interests disclosure

    This study was funded by Eli Lilly and Co. M Ortega, A Molero, S de la Torre, M-H Jen and S Díaz-Cerezo are employees of Eli Lilly and Co. (Spain). M Ortega, A Molero, S de la Torre, M-H Jen and S Díaz-Cerezo hold Eli Lilly and Co. shares. The rest of the authors received an economic compensation from Eli Lilly and Co. for their participation in the study (consultancy and/or data collection tasks). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    The article has been developed with the support of the Clinical Research Organization IQVIA.

    Ethical conduct of research

    The study was approved by the Ethics Committee of Hospital Ramón y Cajal in Madrid. This was an observational study that used data previously collected and did not impose any form of intervention, the data was deidentified to protect subject privacy. A formal consent to release information form was not required from the Ethics Committee of Hospital Ramón y Cajal in Madrid.

    Open Access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest

    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. 68(6), 394–424 (2018).
    • 2. Rawla P, Barsouk A. Epidemiology of gastric cancer: global trends, risk factors and prevention. Prz. Gastroenterol. 14(1), 26–38 (2019).
    • 3. Kamiya S, Rouvelas I, Lindblad M, Nilsson M. Current trends in gastric cancer treatment in Europe. J. Cancer Metastasis Treat. 4(7), 35 (2018).
    • 4. Lopez-Abente G, Nuuez O, Perez-Gomez B et al. La situación del cáncer en España: informe 2015 [the cancer situation in Spain: report 2015]. SSRN Electron J. (2016).
    • 5. Karimi P, Islami F, Anandasabapathy S et al. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol. Biomarkers Prev. 23(5), 700–713 (2014).
    • 6. Crew KD, Neugut AI. Epidemiology of gastric cancer. World J. Gastroenterol. 12(3), 354–362 (2006).
    • 7. Smyth EC, Verheij M, Allum W et al. Gastric cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 27(August), v38–v49 (2016).
    • 8. Kanagavel D, Fedyanin M, Tryakin A, Tjulandin S. Second-line treatment of metastatic gastric cancer: current options and future directions. World J. Gastroenterol. 21(41), 11621–11635 (2015).
    • 9. DIgklia A, Wagner AD. Advanced gastric cancer: current treatment landscape and future perspectives. World J. Gastroenterol. 22(8), 2403–2414 (2016).
    • 10. Cotes Sanchís A, Gallego J, Hernandez R et al. Second-line treatment in advanced gastric cancer: data from the Spanish AGAMENON registry. PLoS ONE 15 (7), e0235848 (2020).
    • 11. Park SH, Lee WK, Chung M et al. Quality of life in patients with advanced gastric cancer treated with second-line chemotherapy. Cancer Chemother. Pharmacol. 57(3), 289–294 (2006).
    • 12. Fuchs CS, Tomasek J, Yong CJ et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): An international, randomised, multicentre, placebo-controlled, Phase 3 trial. Lancet. 383(9911), 31–39 (2014). • This international Phase III clinical trial revealed increased survival of advanced gastric cancer and gastroesophageal juntion adenocarcinoma patients treated with ramucirumab in monotherapy versus placebo and validated for the first-time VEGFR-2 signaling as a therapeutic target in this disease.
    • 13. Wilke H, Muro K, van Cutsem E et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastroesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomized Phase III trial. Lancet Oncol. 15(11), 1224–1235 (2014). • RAINBOW clinical trial demonstrated the efficacy of ramucirumab in combination with paclitaxel versus paclitaxel alone as a second-line treatment for advanced gastric cancer and gastroesophageal junction adenocarcinoma patients.
    • 14. Di Bartolomeo M, Niger M, Tirino G et al. Ramucirumab as second-line therapy in metastatic gastric cancer: real-world data from the RAMoss study. Target Oncol. 13(2), 227–234 (2018). • RAMoss constitutes the first study conducted in Europe which evidenced the safety and effectiveness of ramucirumab in patients included in a compassionate-use program in Italy.
    • 15. Paulson AS, Hess LM, Liepa AM et al. Ramucirumab for the treatment of patients with gastric or gastroesophageal junction cancer in community oncology practices. Gastric Cancer (2018).
    • 16. Jung M, Ryu MH, Oh DY et al. Efficacy and tolerability of ramucirumab monotherapy or in combination with paclitaxel in gastric cancer patients from the Expanded Access Program Cohort by the Korean Cancer Study Group (KCSG). Gastric Cancer 21(5), 819–830 (2018).
    • 17. Eisenhauer EA, Therasse P, Bogaerts J et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45(2), 228–247 (2009).
    • 18. Oken MM, Creech RH, Davis TE. Toxicology and response criteria of the Eastern Cooperative Oncology Group. Am. J. Clin. Oncol. Cancer Clin. Trials 5(6), 649–655 (1982).
    • 19. Jeong O, Jung MR, Ryu SY. Impact of various types of comorbidities on the outcomes of laparoscopic total gastrectomy in patients with gastric carcinoma. J. Gastric Cancer 18(3), 253–263 (2018).
    • 20. Lorenzen S, Stahl M, Hofheinz RD et al. Influence of taxanes on treatment sequence in gastric cancer. Oncol. Res. Treat. 43(1–2), 42–47 (2020).
    • 21. Thuss-Patience PC, Kretzschmar A, Bichev D et al. Survival advantage for irinotecan versus best supportive care as second-line chemotherapy in gastric cancer – a randomised Phase III study of the Arbeitsgemeinschaft Internistische Onkologie (AIO). Eur. J. Cancer 47(15), 2306–2314 (2011).
    • 22. Ford HER, Marshall A, Bridgewater JA et al. Docetaxel versus active symptom control for refractory oesophagogastric adenocarcinoma (COUGAR-02): an open-label, Phase 3 randomised controlled trial. Lancet Oncol. 15(1), 78–86 (2014).
    • 23. Tehfe M, Tabchi S, Laterza MM, Vita De F. Ramucirumab in HER-2-positive gastroesophageal adenocarcinoma: an argument for overcoming trastuzumab resistance. Future Oncol. 14(3), 223–228 (2018).
    • 24. Cyramza: EPAR – risk-management-plan summary (2020). www.ema.europa.eu/en/documents/rmp-summary/cyramza-epar-risk-management-plan-summary_en.pdf