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-world ramucirumab and immune checkpoint inhibitor sequences in US patients with advanced gastroesophageal cancer

    Astra M Liepa

    *Author for correspondence: Tel.: +1 317 277 5623;

    E-mail Address: liepa_astra_m@lilly.com

    Eli Lilly & Company, Lilly Corporate Center, Indianapolis, IN 46285, USA

    ,
    Zhanglin Lin Cui

    Eli Lilly & Company, Lilly Corporate Center, Indianapolis, IN 46285, USA

    ,
    Julie K Beyrer

    Eli Lilly & Company, Lilly Corporate Center, Indianapolis, IN 46285, USA

    ,
    Elizabeth L Hadden

    Eli Lilly & Company, Lilly Corporate Center, Indianapolis, IN 46285, USA

    DeLisle Associates LTD, Cranbrook DR, Indianapolis, IN 46250, USA

    ‡Former employee of DeLisle Associates LTD

    Search for more papers by this author

    &
    Anindya Chatterjee

    Eli Lilly & Company, Lilly Corporate Center, Indianapolis, IN 46285, USA

    Published Online:https://doi.org/10.2217/fon-2022-0604

    Abstract

    Aim: To describe real-world treatment sequences of ramucirumab relative to immune checkpoint inhibitors (ICIs) in patients with advanced gastroesophageal cancer. Methods: Retrospective, observational study including adult patients treated with ramucirumab (April 2014–June 2020) from a nationwide health-record database. Results: In 1117 eligible patients, ramucirumab + paclitaxel was the most common ramucirumab-containing regimen (72.0%). A total of 217 patients also received an ICI. For ramucirumab then ICI (n = 148) and ICI then ramucirumab (n = 50), ramucirumab + taxane and ICI monotherapy were the most frequent approaches, most commonly observed as second- and third-line (2L and 3L). Median time on ramucirumab in 2L and 3L was similar regardless of sequence with ICI. Conclusion: Most patients with advanced gastroesophageal cancer received ramucirumab before ICI, with ramucirumab + paclitaxel as the most common ramucirumab-based regimen.

    Plain language summary

    What is the Order of New Treatments for Gastroesophageal Cancers in the Real World?

    What is this summary about?

    Gastroesophageal cancers (cancers of the stomach or food pipe) which cannot be cured are first treated using traditional chemotherapy. Newer anti-cancer therapies with fewer side effects, such as ramucirumab (RAM) and immune checkpoint inhibitors (ICI), are now available either alone or in combination with chemotherapy. We designed this study to describe the order of use for RAM and ICI.

    What were the results?

    The patients in this study were from Flatiron Health database, which includes electronic medical record data of US patients with gastroesophageal cancers. The included patients had been treated with RAM and were grouped based on the treatments received and in the order in which they received RAM and ICI. Of the patients who received both RAM and ICI, RAM then ICI was the most common order, followed by ICI then RAM and then RAM plus ICI at the same time. RAM in combination with paclitaxel (a chemotherapy) was the most common RAM-containing treatment. The duration of RAM therapy was the same whether patients received the treatment before or after ICI.

    What do results of the study mean?

    These findings can be used by patients with gastroesophageal cancers and oncologists when making treatment decisions, specifically if RAM might be an option when it is time to change treatments. Real-world studies like this help answer questions that were not addressed in clinical trials.

    Gastric and esophageal (and gastroesophageal junction, GEJ) cancers constituted up to 1.4 and 1.0% of the total cancer cases in the USA. More than 60% of the cases were diagnosed at advanced or metastatic stages [1,2]. The five-years survival rates for gastric and esophageal cancers in the USA are 32.4 and 20.0%, respectively [1,2]. Globally, there is an observed trend of decrease in incidence and mortality of gastric and esophageal cancers; however, the mortality due to gastric and esophageal cancers remains high in East Asia compared with other geographies due to higher incidence rates [3,4].

    Chemotherapy is the standard of care for advanced gastroesophageal cancers, with platinum plus fluoropyrimidine-based regimens being the most preferred regimens in first-line (1L) therapy [5,6]. Treatment options beyond the 1L therapy have only been established in the last decade [7–9]. Ramucirumab (RAM), a VEGFR2 inhibitor, was approved by the US FDA as monotherapy or in combination with paclitaxel (PAC) (RAM + PAC) in patients with advanced gastric or GEJ adenocarcinoma previously treated with fluoropyrimidine/platinum [7,10,11]. However, RAM in combination with fluoropyrimidine/platinum is not recommended in 1L gastric or GEJ adenocarcinoma treatment [10]. The development and availability of immune checkpoint inhibitors (ICI) have significantly changed the treatment dynamics of solid cancers, including that of gastroesophageal cancers [12,13]. The findings from clinical studies of pembrolizumab initially led to its approval and use in third-line (3L) or in later for gastric/GEJ adenocarcinoma [14–16], and second-line (2L) esophageal squamous-cell cancer (SCC) [17,18], with nivolumab also approved for 2L esophageal cancer [19,20]. Recently, pembrolizumab [21–23] and nivolumab [9,24] were approved as 1L regimens in combination with chemotherapy for gastroesophageal cancers [21,24] or chemotherapy plus trastuzumab [22] for patients with HER2-positive gastroesophageal adenocarcinoma. With multiple studies demonstrating the benefits of ICI in combination with chemotherapy in gastroesophageal cancer, these combinations are expected to become a new standard of care in 1L therapy [25].

    In addition to chemotherapy options, the National Comprehensive Care Network (NCCN) guidelines recommend RAM ± chemotherapy as 2L regimen for gastroesophageal adenocarcinomas, with ICIs recommended in 2L therapy for microsatellite instability-high (MSI-H) and tumor mutational burden (TMB) high gastroesophageal adenocarcinoma (pembrolizumab), esophageal SCC (nivolumab), and SCC with elevated PD-L1 expression (pembrolizumab) [5,6]. Despite studies demonstrating the vital role of sequence of treatments in clinical outcomes of gastric cancer [26,27], there is no consensus regarding the preferred treatment regimen in the 2L and 3L+ therapy settings in the USA [26].

    Studies in Asia have demonstrated that patients treated with RAM + PAC after failing an ICI treatment had better outcomes versus patients treated with RAM + PAC therapy who were ICI-treatment naive [28,29]. Another study by Kawai et al. demonstrated that 3L RAM monotherapy has similar efficacy and toxicity as that of 2L RAM monotherapy in patients with advanced gastric cancer [30]. In another study, differences in disease characteristics and management of advanced gastric cancer by geography were reported [31]. There are limited prospective clinical trial data and real-world studies available on the sequence and outcomes of RAM and ICI treatments in the USA. Furthermore, there are no real-world data available for potential use of ICI in 1L therapy, including subsequent lines of therapy and outcomes. Therefore, real-world evidence on utilization of RAM before or after ICI initiation in patients with advanced gastroesophageal cancer could contribute to improving evidence-based clinical decision making in the US. This study aimed to fill this evidence gap by describing the sequences of RAM relative to ICI in USA patients receiving treatment for advanced gastroesophageal cancer, including RAM by line of therapy and by regimen, as part of their routine cancer care.

    Patients & methods

    Study design & data source

    This retrospective, observational study used the Advanced Gastric/Esophageal Cancer Cohort from the USA nationwide Flatiron Health electronic health record (EHR)-derived de-identified database, which is a longitudinal database, comprising patient-level structured and unstructured data, curated via technology-enabled abstraction [32,33]. Regimens and lines of therapy in the database were derived using rules based on clinical expert input. The study was conducted using data on patients diagnosed with advanced gastric, GEJ or esophageal cancer from 1 January 2011 through end of follow-up (last visit dates) through 30 June 2020. During the study period, the de-identified data originated from approximately 280 cancer clinics (∼800 sites of care).

    The study followed the ethical principles of the Declaration of Helsinki, Good Pharmacoepidemiology Practices, and applicable laws, and regulations in the USA. These research activities are covered in Flatiron Health's parent protocol which was reviewed and approved by the Copernicus Group Institutional Review Board (IRB). However, this general protocol does not cover targeted research questions or complex statistical analyses. All data were de-identified to protect patients' confidentiality in compliance with Health Insurance Portability and Accountability Act regulations. Informed consent from patients was not required.

    Patient population

    The database included patients diagnosed with advanced gastric, GEJ or esophageal cancer on or after 1 January 2011 and who have ≥2 visits on different days documented in the EHR during that same period. The patients were identified based on the International Classification of Diseases (ICD). For gastric cancer (ICD-9 151.x or ICD-10 C16.x), patients should have had advanced disease at diagnosis with either distant recurrence or a second locoregional recurrence, or a first locoregional recurrence that was not completely resected, or no surgical resection of the primary tumor. For GEJ or esophageal cancer (ICD-9 150.x or ICD-10 C15.x), patients should have had distant metastases at diagnosis or distant recurrence, any locoregional recurrence, or no surgical resection of the primary tumor.

    Study-specific inclusion criteria were: i) initiating 1L therapy for advanced gastric, GEJ or esophageal cancer at age ≥18 years; ii) receiving RAM in any line of therapy on or after 21 April 2014 (initial regulatory approval for RAM) and iii) had structured activity within 90 days of advanced diagnosis date in the EHR.

    Assessments

    We described the treatment sequence with RAM in patients with advanced gastroesophageal cancer as four mutually exclusive patient groups based on the use of the first treatment in the treatment sequence: 1) RAM then ICI, 2) ICI then RAM, 3) RAM with ICI in the same line of therapy and 4) RAM without ICI in any line of therapy. In addition, we described the use of RAM in the sequences by line of therapy and by regimen, patient and disease characteristics, the use of prior therapies by line of therapy (regimens), and the use of subsequent therapies by line of therapy (percentage who receive, regimens, number of subsequent lines). Time on RAM within treatment sequences (duration of treatment) by regimen and by line of therapy was also assessed.

    Patient groups

    Patients in RAM without ICI group were further classified based on the line of therapy: 1L, 2L, 3L and any other line of therapy. Patients were also classified based on RAM use in these sequences by line of therapy (as 1L, 2L, 3L, fourth line or higher [4L+] therapy when either RAM or ICI was used for the first time), by regimen (RAM + PAC, monotherapy, with other agents), and primary tumor site (gastric, GEJ, esophageal). Line of therapy was based on Flatiron Health oncologist-defined rules with a revision so that substitution of any biosimilar agents did not advance the line of therapy.

    Statistical analysis

    Data for the study cohort and subgroups were summarized descriptively. Continuous variables were summarized using mean (standard deviation, SD), median (interquartile range, [IQR]), and range (minimum and maximum). Categorical variables were summarized by patient count and percentage for each category including missing/unknown as a special category. Sankey diagrams were generated to visualize treatment sequences.

    Kaplan–Meier method was used to assess time to event variables, and medians (interquartile range, IQR) were reported. Time on RAM for a line of therapy was calculated using Kaplan–Meier method and was defined as time from the first date of RAM administration to the last date of administration. The time at risk for therapy discontinuation for both discontinued and censored patients was calculated from the first date of the therapy administration to last date of the therapy administration. A discontinuation event was defined by the presence of a subsequent line of therapy, patient death, or any gap of ≥42 days with no RAM treatment before the patient's last activity date; otherwise, the patient was censored on the last administration date for RAM.

    All summaries and analyses were performed using the Instant Health Data (IHD) software (Panalgo, MA, USA) and R, version 3.2.1 (R Foundation for Statistical Computing, Vienna, Austria).

    Results

    Demographics & clinical characteristics

    A total of 10,138 patients comprised the Flatiron Health Advanced Gastric/Esophageal Cancer Cohort, of which, 1117 patients were included in this study, Supplementary Figure 1. Of the 1117 patients, 74.9% were males, 66.0% were Caucasians, mean (SD) age was 63.2 (11.2) years at initiation of 1L therapy and mean (SD) age was 64.0 (11.2) years at initiation of the 1L therapy containing RAM. The majority of the patients (91.6%) in the study were from community oncology settings, with the remainder from the academic setting. Among the patients, 217 received RAM and ICI: RAM before ICI (n = 148, 68.2%), ICI then RAM (n = 50, 23.0%), RAM and ICI in same line of therapy (n = 19, 8.8%). ICI was not administered in any line of therapy for the remaining 900 patients.

    Table 1 presents the baseline and clinical characteristics for each group. Among the patients from different groups, patients in the ICI then RAM group were the oldest (mean [SD] age 65.0 [10.2] years) at 1L initiation and had the highest proportion of patients aged ≥65 years (58.0%). A similar pattern was observed for age at RAM initiation; patients in the ICI then RAM group were older on average (mean [SD] age 66.3 [10.2] years), and patients in RAM with ICI group had the highest proportion of patients aged ≥65 years (68.4%). In all the groups, the majority of the patients had stage IV disease at initial diagnosis and had Eastern Cooperative Oncology Group (ECOG) status of 0 or 1 at 1L therapy initiation. At RAM initiation, RAM then ICI and ICI then RAM groups had >50% patients with ECOG PS of 0 or 1.

    Table 1. Demographics and baseline clinical characteristics.
    CharacteristicRAM then ICI, N = 148ICI then RAM, N = 50RAM with ICI, N = 19RAM without ICI, N = 900
    Age at 1L initiation, year, mean (SD)62.7 (11.3)65.0 (10.2)58.1 (15.5)63.3 (11.1)
    Age group at 1L initiation, year, n (%)    
      <6581 (54.7)21 (42.0)13 (68.4)462 (51.3)
      ≥6567 (45.3)29 (58.0)6 (31.6)438 (48.7)
    Age at RAM initiation, year, mean (SD)63.3 (11.4)66.3 (10.2)58.9 (14.9)64.1 (11.1)
    Age group at RAM initiation, year, n (%)    
      <6576 (51.4)19 (38.0)13 (68.4)442 (49.1)
      ≥6572 (48.7)31 (62.0)6 (31.6)458 (50.9)
    Gender, n (%)    
      Male115 (77.7)35 (70.0)13 (68.4)674 (74.9)
      Female33 (22.3)15 (30.0)6 (31.6)226 (25.1)
    Race, n (%)    
      White90 (60.8)37 (74.0)10 (52.6)600 (66.7)
      Black/African–American12 (8.1)≤5≤553 (5.9)
      Asian8 (5.4)≤5≤542 (4.7)
      Other24 (16.2)6 (12.0)≤5128 (14.2)
      Missing/Unknown14 (9.5)≤5≤572 (8.0)
    Geographic region, n (%)    
      South61 (41.2)17 (34.0)9 (47.4)307 (34.1)
      Northeast31 (21.0)11 (22.0)≤5185 (20.6)
      West24 (16.2)12 (24.0)≤5155 (17.2)
      Midwest16 (10.8)6 (12.0)≤5126 (14.0)
      Missing16 (10.8)≤5≤5127 (14.1)
    Practice setting, n (%)    
      Community138 (93.2)47 (94.0)16 (84.2)822 (91.3)
      Academic10 (6.8)3 (6.0)3 (15.8)78 (8.7)
    Primary tumor site, n (%)    
      Gastric58 (39.2)16 (32.0)9 (47.4)365 (40.6)
      GEJ43 (29.1)17 (34.0)4 (21.1)264 (29.3)
      Esophageal47 (31.8)17 (34.0)6 (31.6)271 (30.1)
    Histology, n (%)    
      Adenocarcinoma141 (95.3)46 (92.0)17 (89.5)874 (97.1)
      SCC4 (2.7)4 (8.0)2 (10.5)12 (1.3)
      Other2 (1.4)0010 (1.1)
      Adenosquamous1 (0.7)002 (0.2)
      Unknown/Not documented0002 (0.2)
    Time between any 1L start and last activity/death, months, median (IQR)16.1 (11.8–25.2)19.5 (11.7–27.4)11.2 (5.4–25.2)12.4 (7.5–19.6)
    Time between RAM 1L start and last activity/death, months, median (IQR)8.8 (5.3–15.1)4.2 (1.3–8.2)4.9 (2.5–10.3)3.6 (1.7–7.4)
    Stage at initial diagnosis, n (%)    
      I3 (2.0)1 (2.0)020 (2.2)
      II11 (7.4)10 (20.0)3 (15.8)86 (9.6)
      III26 (17.6)5 (10.0)3 (15.8)164 (18.2)
      IV98 (66.2)31 (62.0)10 (52.6)551 (61.2)
      Unknown10 (6.8)3 (6.0)3 (15.8)79 (8.8)
    Criterion for advanced diagnosis, n (%)    
      Stage IV/IVB97 (65.5)31 (62.0)10 (52.6)548 (60.9)
      Locoregional or distant recurrence25 (16.9)9 (18.0)4 (21.1)174 (19.3)
      Surgery not received – Due to any reason19 (12.8)10 (20.0)5 (26.3)149 (16.6)
      Incomplete resection7 (4.7)0029 (3.2)
    HER2 status at RAM initiation, n (%)    
      Positive24 (16.2)11 (22.0)5 (26.3)165 (18.3)
      Negative113 (76.4)29 (58.0)12 (63.2)618 (68.7)
      Equivocal1 (0.7)1 (2.0)010 (1.1)
      Unknown/No evidence missing10 (6.8)9 (18.0)2 (10.5)107 (11.9)

    1L: First-line; ECOG: Eastern Cooperative Oncology Group; ICI: Immune checkpoint inhibitor; GEJ: Gastroesophageal junction; ICI: Immune checkpoint inhibitor; IQR: Interquartile range; n: Number of patients with observed results; N: Number of patients in the group; RAM: Ramucirumab; SCC: Squamous-cell carcinoma; SD: Standard deviation.

    Treatment characteristics

    The proportion of patients who received RAM in 2L therapy was the highest among those who received RAM then ICI and lowest in those who received ICI then RAM (RAM then ICI: 61.5%; ICI then RAM: 6.0%; RAM with ICI: 26.3%; RAM without ICI: 52.9%). RAM without ICI was administered primarily in 2L (52.9%) and 3L (22.9%) therapies. Irrespective of treatment sequence, RAM + PAC combination was the most common RAM regimen (72.0%) compared with RAM monotherapy (17.0%) and RAM + Other therapy (11.0%). In patients whose 1L initiated from 2011 to 2016, 13.6% received both RAM and ICI (any sequence). After both RAM and ICI were approved, 27.3% of patients who received 1L therapy between 2011 and 2017 received both RAM and ICI.

    Median time to initiate RAM after 1L was the longest in the ICI then RAM group (12.2 months IQR 8.9–19.4 months). For each group, baseline treatment characteristics including the patients treated with RAM-containing therapy, line of therapy containing first RAM, and number of lines of therapy are included in Table 2.

    Table 2. Baseline Clinical and Treatment Characteristics.
    CharacteristicRAM then ICI, N = 148ICI then RAM, N = 50RAM with ICI, N = 19RAM without ICI, N = 900
    ECOG PS at 1L initiation, n (%)    
      041 (27.7)10 (20.0)2 (10.5)184 (20.4)
      157 (38.5)21 (42.0)5 (26.3)310 (34.4)
      29 (6.1)5 (10.0)4 (21.1)102 (11.3)
      3+5 (3.4)1 (2.0)2 (10.5)24 (2.7)
      Unknown36 (24.3)13 (26.0)6 (31.6)280 (31.1)
    ECOG PS at RAM initiation, n (%)    
      030 (20.3)9 (18.0)1 (5.3)122 (13.6)
      163 (42.6)18 (36.0)5 (26.3)299 (33.2)
      217 (11.5)15 (30.0)4 (21.1)182 (20.2)
      3+5 (3.4)2 (4.0)4 (21.1)68 (7.6)
      Unknown33 (22.3)6 (12.0)5 (26.3)229 (25.4)
    RAM in Line of therapy, n (%)    
      1L19 (12.8)08 (42.1)132 (14.7)
      2L91 (61.5)3 (6.0)5 (26.3)476 (52.9)
      3L25 (16.9)24 (48.0)2 (10.5)206 (22.9)
      4L+13 (8.8)23 (46.0)4 (21.1)86 (9.6)
    Maximum number of lines of therapy, n (%)    
      1006 (31.6)100 (11.1)
      211 (7.4)3 (6.0)5 (26.3)337 (37.4)
      348 (32.4)20 (40.0)4 (21.1)284 (31.6)
      447 (31.8)10 (20.0)1 (5.3)101 (11.2)
      530 (20.3)10 (20.0)053 (5.9)
      612 (8.1)7 (14.0)3 (15.8)25 (2.8)
    RAM Regimens, n (%)    
      RAM + PAC123 (83.1)36 (72.0)0645 (71.7)
      RAM monotherapy10 (6.8)6 (12.0)0174 (19.3)
      RAM + Other therapy15 (10.1)8 (16.0)19 (100)81 (9.0)
    Year of 1L initiation, n (%)    
      20110004 (0.4)
      20123 (2.0)0016 (1.8)
      20132 (1.4)0070 (7.8)
      201411 (7.4)3 (6.0)0140 (15.6)
      201520 (13.5)4 (8.0)4 (21.1)175 (19.4)
      201630 (20.3)8 (16.0)2 (10.5)149 (16.6)
      201738 (25.7)10 (20.0)2 (10.5)123 (13.7)
      201823 (15.5)15 (30.0)5 (26.3)113 (12.6)
      201920 (13.5)9 (18.0)6 (31.6)101 (11.2)
      20201 (0.7)1 (2.0)09 (1.0)
    Time from 1L to RAM initiation in months, median (IQR)6.2 (3.0–10.6)12.2 (8.9–19.4)3.3 (0.0–13.2)7.2 (3.4–12.5)

    1L: First-line therapy, ECOG PS: Eastern Cooperative Oncology Group Performance Status, ICI: Immune checkpoint inhibitor; IQR: Interquartile range; L: Line of therapy; n: Number of patients with observed results; N: Number of patients in the group; PAC: Paclitaxel; RAM: Ramucirumab.

    Treatment patterns

    RAM then ICI Group

    Figure 1 presents the treatment sequences in the 148 patients who received RAM then ICI. All patients received at least two lines of therapy, with 137 receiving 3L therapy, 89 receiving 4L therapy and 42 receiving 5L therapy. In this group, 70.3% (n = 104) of patients received ICI immediately after RAM. The most common treatment received was fluoropyrimidine + platinum in 1L therapy (n = 67/148, 45.3%), RAM + taxanes in 2L therapy (n = 72/148, 48.6%), and ICI monotherapy in 3L therapy (n = 61/137, 44.5%).

    Figure 1. Treatment Regimens in Any Line of Therapy – RAM then ICI Group.

    Regimens may include HER2-targeted therapies (e.g., trastuzumab), which are not presented here. Regimen of HER2-targeted monotherapy is included in ‘Other’.

    FOLFIRI: Chemotherapy regimen consisting of leucovorin calcium (calcium folinate), 5-fluorouracil, and irinotecan; LOT: Line of therapy; N: Number of patients in each line of therapy; n: number of patients in each line of therapy; TAS-102: Trifluridine/tripiracil.

    Irrespective of the sequence, the most common treatment regimens were FOLFOX in 1L (n = 43/148, 29.1%), RAM + PAC in 2L (n = 68/148, 46.0%), and pembrolizumab in 3L (n = 44/137, 32.1%), 4L (n = 25/89, 28.1%), and 5L (n = 20/42, 47.6%) therapies. Other prominent treatment regimens were carboplatin + PAC (n = 14, 9.5%), and PAC + RAM (n = 13, 8.8%) in 1L therapy; FOLFOX (n = 14, 9.5%) and pembrolizumab (n = 8, 54.%) in 2L therapy; and PAC + RAM (n = 19, 13.9%) and FOLFIRI (n = 18, 13.1%) in 3L therapy. All other regimens in each line of therapy are presented in Table 3.

    Table 3. Most common regimens in each line of therapy – RAM then ICI group.
    Line of therapy and RegimenRAM then ICI, N = 148
    1L, n (%)148 (100.0)
      FOLFOX43 (29.1)
      Carboplatin + PAC14 (9.5)
      PAC + RAM13 (8.8)
      Capecitabine + epirubicin + oxaliplatin7 (4.7)
      FOLFOX + trastuzumab7 (4.7)
    2L, n (%)148 (100.0)
      PAC + RAM68 (46.0)
      FOLFOX14 (9.5)
      Pembrolizumab8 (5.4)
      Nivolumab5 (3.4)
      FOLFIRI5 (3.4)
    3L, n (%)137 (92.6)
      Pembrolizumab44 (32.1)
      PAC + RAM19 (13.9)
      FOLFIRI18 (13.1)
      Nivolumab17 (12.4)
      Irinotecan5 (3.7)
    4L, n (%)89 (60.1)
      Pembrolizumab25 (28.1)
      Nivolumab12 (13.5)
      FOLFIRI9 (10.1)
      PAC + RAM8 (9.0)
      Irinotecan5 (5.6)
    5L, n (%)42 (28.4)
      Pembrolizumab20 (47.6)
      Nivolumab3 (7.1)
      FOLFOX2 (4.8)
      RAM1 (2.4)
      PAC + RAM + trastuzumab1 (2.4)

    All data are presented as n (%).

    1L: First-line therapy, 2L: Second-line therapy, 3L: Third-line therapy, 4L: Fourth-line therapy, 5L: Fifth-line therapy, FOLFIRI: Folinic acid, 5-fluorouracil, and irinotecan; FOLFOX: Folinic acid, 5-fluorouracil, and oxaliplatin; ICI: Immune checkpoint inhibitor; n: Number of patients with observed results; N: Number of patients in the group; PAC: Paclitaxel; RAM: Ramucirumab.

    There may be additional regimens with the same minimum number of patients which are not presented here.

    ICI then RAM Group

    Figure 2 presents the treatment sequences in the 50 patients who received ICI then RAM. All 50 patients received at least two lines of therapy, with 47 receiving 3L therapy, 27 receiving 4L therapy, and 17 receiving 5L therapy. A total of 84.0% (n = 42) patients received ICI immediately prior to RAM. The most common treatments received in first three lines were fluoropyrimidine + platinum in 1L (n = 26, 52.0%), ICI monotherapy in 2L (n = 27, 54.0%), and RAM + taxane in 3L (n = 19, 40.4%) therapies.

    Figure 2. Treatment Regimens in Any Line of Therapy – ICI then RAM Group.

    Regimens may include HER2-targeted therapies (eg, trastuzumab), which are not presented here.

    FOLFIRI: Chemotherapy regimen consisting of leucovorin calcium (calcium folinate), 5-fluorouracil, and irinotecan; LOT: Line of therapy; N: Number of patients in each line of therapy; TAS-102: Trifluridine/tripiracil.

    In the ICI then RAM group, the most common treatment regimens across lines of therapy were FOLFOX (n = 19/50, 38.0%) in 1L, pembrolizumab (n = 19/50, 38.0%) in 2L, and RAM + PAC (n = 18/47, 38.3%) in 3L. Other prominent treatment regimens for the group included carboplatin + PAC (n = 7/50, 14.0%), and FOLFOX + trastuzumab (n = 6/50, 12.0%) in 1L therapy; nivolumab (n = 7/50, 14.0%) and FOLFOX (n = 4/50, 8.0%) in 2L therapy; and pembrolizumab (n = 11/47, 23.4%) and FOLFIRI (n = 3/47, 6.4%) in 3L therapy (Table 4).

    Table 4. Most common regimens in each line of therapy – ICI then RAM group.
    Line of therapy and RegimenICI then RAM, N = 50
    1L, n (%)50 (100.0)
      FOLFOX19 (38.0)
      Carboplatin + PAC7 (14.0)
      FOLFOX + trastuzumab6 (12.0)
      Fluorouracil + docetaxel + leucovorin + oxaliplatin4 (8.0)
      Pembrolizumab3 (6.0)
    2L, n (%)50 (100.0)
      Pembrolizumab19 (38.0)
      Nivolumab7 (14.0)
      FOLFOX4 (8.0)
      FOLFIRI3 (6.0)
      Carboplatin + PAC3 (6.0)
    3L, n (%)47 (94.0)
      PAC + RAM18 (38.3)
      Pembrolizumab11 (23.4)
      FOLFIRI3 (6.4)
      Trifluridine/tipiracil2 (4.3)
      Nivolumab2 (4.3)
    4L, n (%)27 (54.0)
      PAC + RAM8 (29.6)
      Pembrolizumab4 (14.8)
      RAM3 (11.1)
      FOLFIRI2 (7.4)
      PAC + pembrolizumab + RAM1 (3.7)
    5L, n (%)17 (34.0)
      PAC + RAM8 (47.1)
      FOLFOX2 (11.8)
      Irinotecan1 (5.9)
      Pembrolizumab1 (5.9)
      Docetaxel + irinotecan + RAM1 (5.9)

    All data are presented as n (%).

    1L: First-line therapy, 2L: Second-line therapy, 3L: Third-line therapy, 4L: Fourth-line therapy, 5L: Fifth-line therapy, FOLFIRI: Folinic acid, 5-fluorouracil, and irinotecan; FOLFOX: Folinic acid, 5-fluorouracil, and oxaliplatin; ICI: Immune checkpoint inhibitor; n: Number of patients with observed results; N: Number of patients in the group; PAC: Paclitaxel; RAM: Ramucirumab.

    There may be additional regimens with the same minimum number of patients which are not presented here.

    RAM with ICI Group

    RAM + ICI was mostly received in 1L therapy (n = 8/19, 42.1%), and 2L therapy (n = 6/13, 46.2%). In 3L therapy, 28.6% (n = 2/7) patients each received FOLFIRI/irinotecan or RAM + ICI. Treatment sequences are presented in Supplementary Figure 2.

    Among the treatment regimens in different lines of therapy, the most common regimens were FOLFOX (n = 3/19, 15.8%) in 1L therapy and RAM + PAC + pembrolizumab (n = 4/13, 30.8%) in 2L therapy. Overall, 42.1% patients (n = 8) received 3L therapy, with all patients receiving different treatment regimens, Table 5.

    Table 5. Most common regimens in each line of therapy – RAM with ICI.
    Line of therapy and RegimenRAM with ICI, N = 19
    1L, n (%)19 (100.0)
      FOLFOX3 (15.8)
      Cisplatin + PAC + pembrolizumab + RAM2 (10.5)
      PAC + pembrolizumab + RAM2 (10.5)
      Carboplatin + PAC2 (10.5)
      Pembrolizumab + RAM2 (10.5)
    2L, n (%)13 (68.4)
      PAC + pembrolizumab + RAM4 (30.8)
      Docetaxel + irinotecan + pembrolizumab + RAM1 (7.7)
      CAPEOX1 (7.7)
      Atezolizumab + pembrolizumab + RAM + trastuzumab1 (7.7)
      Fluorouracil + cisplatin + docetaxel + oxaliplatin1 (7.7)
    3L, n (%)8 (42.1)
      Irinotecan1 (12.5)
      Fluorouracil + carboplatin + PAC-protein-bound1 (12.5)
      Docetaxel + pembrolizumab + RAM1 (12.5)
      PAC + pembrolizumab + trastuzumab1 (12.5)
      Irinotecan + pembrolizumab + RAM1 (12.5)
    4L, n (%)4 (21.1)
      Capecitabine1 (25.0)
      Nivolumab + RAM1 (25.0)
      PAC + pembrolizumab + RAM1 (25.0)
      Capecitabine + PAC-protein-bound1 (25.0)
    5L, n (%)3 (15.8)
      CAPEOX + trastuzumab1 (33.3)
      PAC-protein bound + pembrolizumab + RAM1 (33.3)
      FOLFIRI1 (33.3)

    †All regimens for 4L and 5 therapies presented in table.

    All data are presented as n (%).

    1L: First-line therapy, 2L: Second-line therapy, 3L: Third-line therapy, 4L: Fourth-line therapy, 5L: Fifth-line therapy, CAPEOX: Capecitabine and oxaliplatin; FOLFIRI: Folinic acid, 5-fluorouracil, and irinotecan; FOLFOX: Folinic acid, 5-fluorouracil, and oxaliplatin; ICI: Immune checkpoint inhibitor; n: Number of patients with observed results; N: Number of patients in the group; PAC: Paclitaxel; RAM: Ramucirumab.

    There may be additional regimens with the same minimum number of patients which are not presented here.

    RAM without ICI Group

    Supplementary Figure 3 presents the treatment sequences for the 900 patients receiving RAM without ICI. Of these patients, 800 received 2L, 463 received 3L, 179 received 4L, and 78 received 5L therapy. The most common treatments received were fluoropyrimidine + platinum (n = 375/900, 41.7%) in 1L and RAM + taxane (n = 369/800, 46.1%) in 2L.

    Irrespective of treatment sequence, the most common treatment regimen in the RAM without ICI group was FOLFOX in 1L (n = 269/900, 29.9%). RAM + PAC was the most common regimen in 2L (n = 346/800, 43.3%), 3L (n = 137/463, 29.6%), 4L (n = 32/179, 17.9%) and 5L (n = 15/78, 19.2%). Supplementary Tables 1–3 present the treatment regimens for the RAM without ICI group based on the line of therapy in which RAM was received.

    Time on RAM in 2L & 3L therapy

    When RAM was administered in combination with PAC, median duration of RAM therapy did not exceed 3.0 months. Patients who received RAM + PAC in 2L therapy in the RAM then ICI group had a similar duration of therapy compared with those who received 2L RAM + PAC therapy in the RAM without ICI group (3.0 months [IQR, 1.7–5.1 months] vs 2.3 months [IQR 1.1–5.0 months]). Patients who received RAM + PAC in 3L therapy in the RAM then ICI group had a similar median duration of therapy compared with those who received 3L RAM + PAC therapy in the ICI then RAM and the RAM without ICI groups. For patients who received RAM monotherapy in 2L therapy, the duration of RAM therapy in RAM then ICI group was similar compared with those who received 2L RAM monotherapy in the RAM without ICI group. For 3L therapy, duration of RAM monotherapy in RAM then ICI group was similar to ICI then RAM and RAM without ICI groups, Table 6.

    Table 6. Median time on RAM therapy (months) - RAM + PAC and RAM monotherapy.
    TherapyRAM then ICI, N = 148ICI then RAM, N = 50RAM without ICI, N = 900
    RAM + PAC   
      2L, n (IQR)3.0 (1.7–5.1)NA2.3 (1.1–5.0)
      3L, n (IQR)2.8 (1.4–5.2)1.9 (0.5–5.8)2.3 (1.4–4.2)
    RAM monotherapy   
      2L, n (IQR)2.8 (2.3–4.6)2.3 (1.1–5.2)
      3L, n (IQR)1.9 (1.4–2.5)1.3 (0.5–2.0)1.4 (0.5–2.8)

    †No 2L RAM monotherapy.

    Data are presented median (IQR), month, based on Kaplan–Meier estimates, unless specified.

    2L: 2nd line; 3L: 3rd line; ICI: Immune checkpoint inhibitor; IQR: Interquartile range; n: Number of patients with observed results; N: Number of patients in the group; NA: Not estimable; PAC: Paclitaxel; RAM: Ramucirumab.

    The table excluded RAM + ICI in same line group.

    Discussion

    To the best of our knowledge, this is the first study describing the sequence of RAM and ICI therapies in patients with advanced or metastatic gastroesophageal cancer from the USA. The analysis is based on a large USA multi-institutional and geographically diverse dataset. The most common RAM-containing regimen in this study was RAM + PAC and the most common ICI regimen was pembrolizumab monotherapy. Whether RAM was used before or after ICI, treatment most commonly initiated with 1L fluoropyrimidine + platinum therapy, with 1L FOLFOX as the most common regimen.

    In this descriptive analysis, a greater number of patients received RAM then ICI (n = 148) compared with ICI then RAM (n = 50), indicating a greater use of 2L RAM followed by 3L ICI than 2L ICI followed by 3L RAM for the time period of April 2014 to June 2020. While some patients received RAM (most commonly with PAC) in 3L therapy, the most common use of RAM was with PAC in 2L therapy. In addition, RAM monotherapy was used more commonly in later lines of therapy. The treatments administered in 2L therapy were aligned with the NCCN guidelines for gastric and esophageal/GEJ cancers, which also recommend use of RAM + PAC (preferred regimen) or RAM monotherapy (other recommended regimen) in gastric cancer (category 1) and the use of RAM + PAC for adenocarcinoma (preferred regimen: category 1 for GEJ and category 2A for esophageal carcinoma) or RAM monotherapy other recommended regimen: category 1 for GEJ and category 2A for esophageal carcinoma [5,6]. During the study period, NCCN guidelines recommended the use of ICIs for gastroesophageal adenocarcinoma for later lines of therapy, except in cases of MSI-H or TMB high tumors [5,34]. This study did not appreciably include patients with SCC histology as RAM is approved only for gastric/GEJ adenocarcinoma [7]. A systematic review of the treatment regimens used in advanced gastric/GEJ adenocarcinoma synthesized the evidence from 70 publications on clinical studies to provide insights on evidence-based treatment sequence. The review determined the optimal treatments in each line by HER2 expression status and reported that for patients with HER2-negative status, 2L RAM + PAC and 3L ICI therapy was considered as the optimal treatment sequence. However, patients with HER2-negative status and CPS ≥5 or ≥10 were considered eligible for ICI therapy in 1L [26]. The systematic review was limited by the fact that it included only a descriptive cross-trial comparison of the randomized clinical trials and comprised evidence available only at the time of conducting the review (2009–2019) [26], which was before the US FDA approval of 1L ICIs and their incorporation into the NCCN guidelines [8,9,16,23].

    Other studies have demonstrated that exposure to ICI therapies before RAM plus taxane therapy improved the objective response rate (ORR), progression-free survival (PFS) and overall survival (OS) in patients with advanced gastroesophageal adenocarcinoma. A previous study has shown that tumor response to RAM-containing 2L therapy is an independent predictor of OS with ICI in 3L therapy. With 3L ICI, OS was significantly longer in patients who had achieved response with 2L RAM-containing regimen versus patients without response to 2L RAM-containing regimen (p < 0.001) [35]. In a retrospective study of advanced gastroesophageal adenocarcinoma, investigators compared outcomes of 19 patients who had received RAM + PAC after ICI therapy with 68 patients who had received RAM + PAC without preceding ICI therapy. Compared with the ICI-naive group, patients who received RAM + PAC after ICI had better effectiveness outcomes [28]. Another retrospective analysis included 233 patients with advanced gastric cancer: 67 exposed to ICI before RAM and 166 ICI naive. Patients exposed to ICI before RAM + taxane therapy had significantly better effectiveness outcomes compared with the ICI naive group [29]. Better outcomes in patients exposed to ICI therapy before RAM + taxanes was attributed to Tregs, tumor associated macrophages, and enhanced PD-1 inhibitor activity due to persistence of PD-1 blocking antibodies [28,29]. These studies suggest potential enhanced effectiveness for RAM + taxane after prior ICI exposure which could be a sequential option for patients who receive 1L ICI-containing regimens. The current study was not designed to compare effectiveness of sequences; tumor assessments required for ORR and PFS analyses are not included in the database, and the definitions we used to categorize treatment sequences would not be suitable to use in comparative OS analyses. While the sequence of therapy could affect the duration on RAM in each group, no important differences in duration were observed across groups. The median time on RAM + PAC therapy in the RAM then ICI group was similar to the ICI then RAM and RAM without ICI groups in 2L and 3L therapies. Similar observations were made for RAM monotherapy in the three groups for 2L and 3L therapies. Future research is needed to understand the optimal treatment sequence for the patients with advanced gastroesophageal cancer in the USA. As the opportunity to evaluate sequences is limited in the clinical trial setting, real-world studies such as this are a potential source of evidence.

    Our study has several strengths. These included a relatively large, nationwide, multi-institution study design, and timeframe of the study which minimized the potential COVID impact on cancer treatment. The majority of the cohort received a 1L platinum-based regimen, representative of the standard of care for advanced gastroesophageal cancers. There are a few limitations to our research. First, the derived lines of therapy may not accurately represent all the treatment regimens and sequences. Second, the lack of ICI biomarker data in the Flatiron Health database at the time of this analysis limits the characterization of ICI use. Third, although we did not analyze surgical history, the majority of patients were initially diagnosed with advanced disease at baseline. A previous study utilizing the same database reported resection in <15% patients, thus, limiting any impact of perioperative chemotherapy [36]. Fourth, the results are descriptive with no formal comparisons between groups. Last, the study period does not include the recent introduction of 1L ICI, so the data may not reflect the most current treatment paradigm. However, until real-world data reflecting 1L ICI are available, data from this study suggest RAM effectiveness regardless of sequence.

    Conclusion

    In real-world routine practice in the USA during April 2014 through June 2020, patients with advanced gastric, esophageal, or GEJ cancer who received both RAM and ICI typically received RAM before an ICI, with RAM most commonly administered in 2L therapy. Among the different RAM-containing regimens, RAM + PAC was the most common RAM regimen irrespective of line of therapy or the sequence of administration of RAM, i.e., RAM before or after ICI. The study also showed that time on RAM was similar irrespective of the sequence. With approval of ICIs as 1L therapy, the treatment sequences are likely to evolve.

    Summary points
    • This study analyzed patient data in a nationwide, real-world, electronic health record database treated with ramucirumab (RAM) between April 2014 and June 2020 (N = 1117).

    • Patients were grouped by treatment sequence: RAM then immune checkpoint inhibitor (ICI) (n = 148), ICI then RAM (n = 50), RAM with ICI (n = 19), and RAM without ICI (n = 900).

    • We presented the most common treatment approaches and most common drugs used in the four groups starting with their first line of therapy through up to the fifth line.

    • The most common RAM regimen was in combination with paclitaxel.

    • We observed that regardless of sequence and line of therapy, time on ramucirumab was similar.

    • When patients with advanced gastroesophageal cancer received both RAM and ICI, most patients received the sequence of RAM before ICI, but time on RAM therapy was similar irrespective of sequence.

    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-2022-0604

    Author contributions

    All authors made a significant contribution to the work reported whether that is in the conception, study design, execution, acquisition of data, analysis, and interpretation, or in all these areas; took part in drafting, revising, or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for the content of the article.

    Financial & competing interests disclosure

    This study sponsored by Eli Lilly and Company. Astra M. Liepa, Zhanglin Lin Cui, Julie K. Beyrer, and Anindya Chatterjee are employees and stockholders at Eli Lilly and Company. Elizabeth L. Hadden is a former employee of DeLisle Associates LTD, and was sub-contracted by Eli Lilly and Company at the time of manuscript development. 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 authors would like to thank Karan Sharma from Eli Lilly and Company for providing medical writing and editorial support.

    Ethical conduct of research

    The study followed the ethical principles of the Declaration of Helsinki, Good Pharmacoepidemiology Practices, and applicable laws, and regulations in the US. These research activities are covered in Flatiron Health's parent protocol which was reviewed and approved by the Copernicus Group Institutional Review Board (IRB). However, this general protocol does not cover targeted research questions or complex statistical analyses. All data were de-identified to protect patients' confidentiality in compliance with Health Insurance Portability and Accountability Act regulations. Informed consent from patients was not required.

    Data sharing statement

    The data that support the findings of this study have been originated by Flatiron Health, Inc. These de-identified data may be made available upon request, and are subject to a license agreement with Flatiron Health; interested researchers should contact to determine licensing terms.

    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; •• of considerable interest

    References

    • 1. Surveillance, Epidemiology, and End Results Program. Cancer Stat Facts: Stomach Cancer. Available on: https://seer.cancer.gov/statfacts/html/stomach.html (Accessed on 17 January 2022).
    • 2. Surveillance, Epidemiology, and End Results Program. Cancer Stat Facts: Esophageal Cancer. Available on: https://seer.cancer.gov/statfacts/html/esoph.html (Accessed on 17 January 2022).
    • 3. GBD 2017 Stomach Cancer Collaborators. The global, regional, and national burden of stomach cancer in 195 countries, 1990–2017: a systematic analysis for the Global Burden of Disease study 2017. Lancet Gastroenterol. Hepatol. 5(1), 42–54 (2020).
    • 4. GBD 2017 Oesophageal Cancer Collaborators. The global, regional, and national burden of oesophageal cancer and its attributable risk factors in 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol. Hepatol. 5, 582–597 (2020).
    • 5. National Comprehensive Care Network. Clinical Practice Guidelines in Oncology (NCCN Guidelines). Gastric Cancer Version 2.2022. Available on: www.nccn.org/professionals/physician_gls/pdf/gastric.pdf (Accessed on 17 Jan 2022). • This is an updated guidelines for clinical practice in gastro-oncology
    • 6. National Comprehensive Care Network. Clinical Practice Guidelines in Oncology (NCCN Guidelines). Esophageal and Esophageal Junction Cancers Version 2.2022. Available on: www.nccn.org/guidelines/guidelines-detail?category=1&id=1433 (Accessed on 30 March 2022). •• This is an updated guidelines for clinical practice in gastro-oncology.
    • 7. Casak SJ, Fashoyin-Aje I, Lemery SJ et al. FDA approval summary: ramucirumab for gastric cancer. Clin. Cancer Res. 21(15), 3372–3376 (2015).
    • 8. FDA grants accelerated approval to pembrolizumab for HER2-positive gastric cancer. Available on: www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-pembrolizumab-her2-positive-gastric-cancer (Accessed on 17 January 2022).
    • 9. FDA approves nivolumab in combination with chemotherapy for metastatic gastric cancer and esophageal adenocarcinoma. Available on: www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-nivolumab-combination-chemotherapy-metastatic-gastric-cancer-and-esophageal (Accessed on 17 January 2022).
    • 10. Fuchs CS, Shitara K, Di Bartolomeo M et al. Ramucirumab with cisplatin and fluoropyrimidine as first-line therapy in patients with metastatic gastric or junctional adenocarcinoma (RAINFALL): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 20(3), 420–435 (2019).
    • 11. Wilke H, Muro K, Van Cutsem E et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol. 15(11), 1224–1235 (2014).
    • 12. Robert C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat. Commun. 11(1), 3801 (2020).
    • 13. Zhang Z, Xie T, Zhang X, Qi C, Shen L, Peng Z. Immune checkpoint inhibitors for treatment of advanced gastric or gastroesophageal junction cancer: current evidence and future perspectives. Chin. J. Cancer Res. 32(3), 287–302 (2020).
    • 14. Fuchs CS, Doi T, Jang RW et al. Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: phase 2 clinical KEYNOTE-059 trial. JAMA Oncol. 4(5), e180013 (2018).
    • 15. FDA grants accelerated approval to pembrolizumab for advanced gastric cancer. Available on: www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-pembrolizumab-advanced-gastric-cancer (Accessed on 15 February 2022).
    • 16. FDA approves pembrolizumab for esophageal or GEJ carcinoma. Available on: www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-esophageal-or-gej-carcinoma (Accessed on 15 February 2022).
    • 17. Kojima T, Shah MA, Muro K et al. Randomized phase III KEYNOTE-181 study of pembrolizumab versus chemotherapy in advanced esophageal cancer. J. Clin. Oncol. 38(35), 4138–4148 (2020).
    • 18. FDA approves pembrolizumab for advanced esophageal squamous cell cancer. Available on: www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-advanced-esophageal-squamous-cell-cancer (Accessed on 2 March 2022).
    • 19. Kato K, Cho BC, Takahashi M et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 20(11), 1506–1517 (2019).
    • 20. FDA approves nivolumab for esophageal squamous cell carcinoma. Available on: www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-nivolumab-esophageal-squamous-cell-carcinoma (Accessed on 2 March 2022).
    • 21. Sun JM, Shen L, Shah MA et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet 398(10302), 759–771 (2021).
    • 22. Janjigian YY, Kawazoe A, Yanez P et al. The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer. Nature 600(7890), 727–730 (2021).
    • 23. FDA D.I.S.C.O. Burst Edition: FDA approval of Keytruda (pembrolizumab) in combination with trastuzumab, fluoropyrimidine- and platinum-containing chemotherapy for the first-line treatment of patients with locally advanced unresectable or metastatic HER2 positive gastric or gastroesophageal junction adenocarcinoma. Available on: www.fda.gov/drugs/resources-information-approved-drugs/fda-disco-burst-edition-fda-approval-keytruda-pembrolizumab-combination-trastuzumab-fluoropyrimidine (Accessed on 15 February 2022).
    • 24. Janjigian YY, Shitara K, Moehler M et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet 398(10294), 27–40 (2021).
    • 25. Paydary K, Reizine N, Catenacci DVT. Immune-checkpoint inhibition in the treatment of gastro-esophageal cancer: a closer look at the emerging evidence. Cancers (Basel) 13(23), 5929 (2021).
    • 26. Catenacci DV, Chao J, Muro K et al. Toward a treatment sequencing strategy: a systematic review of treatment regimens in advanced gastric cancer/gastroesophageal junction adenocarcinoma. Oncologist 26(10), e1704–e1729 (2021).
    • 27. Li SS, Klempner SJ, Costantino CL et al. Impact of treatment sequencing on survival for patients with locally advanced gastric cancer. Ann. Surg. Oncol. 28(5), 2856–2865 (2021).
    • 28. Kankeu Fonkoua LA, Chakrabarti S, Sonbol MB et al. Outcomes on anti-VEGFR-2/paclitaxel treatment after progression on immune checkpoint inhibition in patients with metastatic gastroesophageal adenocarcinoma. Int. J. Cancer 149(2), 378–386 (2021). •• This study suggests that outcomes on immune checkpoint inhibitors (ICI)-experienced patients appear to be better than expected with anti-VEGFR-2/paclitaxel treatment.
    • 29. Sasaki A, Kawazoe A, Eto T et al. Improved efficacy of taxanes and ramucirumab combination chemotherapy after exposure to anti-PD-1 therapy in advanced gastric cancer. ESMO Open 4(Suppl. 2), e000775 (2020). •• This study suggests that outcomes on ICI-experienced patients appear to be better than expected with anti-VEGFR-2/paclitaxel treatment.
    • 30. Kawai S, Fukuda N, Yamamoto S et al. Retrospective observational study of salvage line ramucirumab monotherapy for patients with advanced gastric cancer. BMC Cancer 20(1), 338 (2020). •• This study suggests that outcomes on ICI-experienced patients appear to be better than expected with anti-VEGFR-2/paclitaxel treatment.
    • 31. Kim R, Tan A, Choi M, El-Rayes BF. Geographic differences in approach to advanced gastric cancer: is there a standard approach? Crit. Rev. Oncol. Hematol. 88(2), 416–426 (2013).
    • 32. Ma X, Long L, Moon S, Adamson BJS, Baxi SS. Comparison of population characteristics in real-world clinical oncology databases in the US: Flatiron Health, SEER, and NPCR. MedRxiv, https://doi.org/10.1101/2020.03.16.20037143 (2020).
    • 33. Birnbaum B, Nussbaum N, Seidl-Rathkopf K et al. Model-assisted cohort selection with bias analysis for generating large-scale cohorts from the EHR for oncology research. ArXiv., Doi: 10.48550/arXiv.2001.09765 (13 January 2020). Available on: https://arxiv.org/abs/2001.09765
    • 34. National Comprehensive Care Network. Clinical Practice Guidelines in Oncology (NCCN Guidelines). Esophageal and Esophageal Junction Cancers Version 1.2022. Available on: www.nccn.org/professionals/physician_gls/pdf/esophageal.pdf (Accessed on 17 January 2022).
    • 35. Kim J, Byeon S, Kim H et al. Impact of prior ramucirumab use on treatment outcomes of checkpoint inhibitors in advanced gastric cancer patients. Target Oncol. 15(2), 203–209 (2020).
    • 36. Barzi A, Hess LM, Zhu YE et al. Real-world outcomes and factors associated with the second-line treatment of patients with gastric, gastroesophageal junction, or esophageal adenocarcinoma. Cancer Control 26(1), Doi: 10.1177/1073274819847642 (2019).