Your activity: 42 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Progressive, locally advanced unresectable, and metastatic esophageal and gastric cancer: Approach to later lines of systemic therapy

Progressive, locally advanced unresectable, and metastatic esophageal and gastric cancer: Approach to later lines of systemic therapy
Author:
Harry H Yoon, MD, MHS
Section Editor:
Richard M Goldberg, MD
Deputy Editor:
Diane MF Savarese, MD
Literature review current through: Dec 2022. | This topic last updated: Jul 13, 2022.

INTRODUCTION — Cancers of the upper gastrointestinal tract are highly lethal malignancies. Locally advanced, unresectable, and metastatic esophagogastric cancers are not curable, and the goals of therapy include symptom palliation and prolongation of survival. Palliative modalities for advanced esophageal or gastric cancer can be either local or systemic. While systemic therapy is the most effective treatment modality for patients with metastatic disease and it may adequately palliate dysphagia and other symptoms, such as nausea, pain, obstruction, perforation, or bleeding, from a locally advanced or locally recurrent primary tumor, they often require multidisciplinary management using endoscopic, surgical, radiotherapeutic, or other approaches. (See "Local palliation for advanced gastric cancer" and "Endoscopic palliation of esophageal cancer".)

This topic review will cover the approach to later lines of systemic therapy in patients who fail initial systemic therapy for advanced unresectable, and metastatic esophageal and gastric cancer. The choice of initial systemic treatment for locally advanced and metastatic disease, local methods for palliation in patients with advanced disease, and hepatic metastasectomy for the rare patient with isolated gastric cancer liver metastases are discussed separately. (See "Initial systemic therapy for locally advanced unresectable and metastatic esophageal and gastric cancer" and "Local palliation for advanced gastric cancer" and "Endoscopic palliation of esophageal cancer" and "Surgical management of invasive gastric cancer", section on 'Metastasectomy'.)

APPROACH TO THE PATIENT

Goals of therapy — As with first-line therapy, the goals of second-line therapy for metastatic esophageal and gastric cancer are to palliate symptoms and improve survival:

A survival benefit for second-line cytotoxic chemotherapy has been shown in at least five randomized trials of ramucirumab (both alone and with paclitaxel), single-agent docetaxel, and irinotecan [1-5].

Furthermore, a Cochrane review of salvage systemic therapy for advanced gastric and esophagogastric junction (EGJ) adenocarcinoma after failure of a first-line platinum/fluoropyrimidine-containing regimen concluded that both chemotherapy and immunotherapy using immune checkpoint inhibitors improve survival over placebo in the second-line setting [6].

Patient selection — We do not use any specific algorithm to select patients for second-line treatment. Instead, we make this decision on a case-by-case basis, taking into account performance status, comorbidity, goals of care, patient preference, the overall medical condition, and disease status. For patients with a poor performance status, poorly controlled comorbidity, or a preference for no additional therapy, we suggest supportive care alone rather than second-line therapy.

The criteria to select patients for second-line systemic therapy have not been established. In one study, five factors were identified that were independently associated with poor survival: a performance status of 2, hemoglobin ≤11.5 g/dL, serum carcinoembryonic antigen (CEA) level >50 ng/mL, three or more metastatic sites, and a time to progression of six or fewer months after the first-line regimen [7]. A prognostic index was developed, which divided patients into low- (no risk factors), intermediate- (one or two risk factors), or high-risk (three or more risk factors) groups with different median survival durations (12.7, 7.1, and 3.3 months, respectively). Although this model is prognostic, none of these factors can be used to predict which patients might benefit from a second-line regimen.

Multipanel somatic and germline genomic testing — Somatic (tumoral) and germline genomic testing in order to identify potentially actionable targets is indicated for patients who might be eligible for immunotherapy or molecularly targeted therapy.

The American Society of Clinical Oncology has issued a provisional clinical opinion that supports somatic and germline genomic testing in metastatic or advanced cancer when there are genomic biomarker-linked therapies approved by regulatory agencies for that type of cancer [8]. Given the tissue-agnostic approvals for any advanced cancer with a high tumor mutational burden (TMB) or DNA mismatch repair deficiency (checkpoint inhibitor immunotherapy), or neurotrophic tyrosine receptor kinase (NTRK) fusions (TRK inhibitors), this provides a rationale for testing for all solid tumors, if the individual would be a candidate for these treatments. For patients without an approved genomic biomarker-linked therapy, testing should be considered to determine candidacy for targeted therapies approved for other diseases; however, off-label/off-study use of such therapies is not recommended when a clinical trial is available, or without evidence of meaningful efficacy in clinical trials. We agree with this guidance.

Regimen choice — Patient preference, quality of life, histologic type, the results of genomic testing, and the specific regimen that was used for initial therapy are key considerations when choosing the therapeutic approach for later lines of therapy. For patients who retain an adequate performance status after the first-line regimen there is no standard approach to second-line therapy and beyond. Particularly in view of the lack of data showing benefit of multiagent cytotoxic chemotherapy versus monotherapy [9,10], we emphasize quality of life and minimization of side effects when choosing the regimen.

For most patients, we prefer enrollment in a clinical trial if available. If trials are unavailable or patients are ineligible, our general approach is summarized below:

Immunotherapy – Immunotherapy targeting programmed cell death 1 (PD-1) is an option for selected patients.

In many cases, PD-1 targeted therapy will have been administered with the first-line chemotherapy regimen. We would generally not pursue anti-PD-1 immunotherapy for later lines of therapy if disease progression occurred during prior PD-1 targeted therapy given in the first-line setting. Rechallenge may be considered, preferably in the context of a clinical trial, if prior anti-PD-1 immunotherapy was discontinued due to a specific toxicity that has since resolved, and possibly for patients with progression at few sites (oligoprogression) after a prior objective response or prolonged period of stable disease during initial PD-1 targeted immunotherapy. (See "Initial systemic therapy for locally advanced unresectable and metastatic esophageal and gastric cancer", section on 'Front-line immunotherapy'.)

The following recommendations apply to patients who did not have progressive disease during front-line immunotherapy:

Pembrolizumab is an option for second-line therapy in patients whose tumors have a deficiency in mismatch repair (dMMR) or high levels of TMB if they did not receive front-line immunotherapy. (See 'Defective mismatch repair' below and 'High tumor mutational burden' below.)

For squamous cell cancers, pembrolizumab is also an option for those with high levels of programmed cell death ligand 1 (PD-L1) expression (combined positive score [CPS] ≥10), and nivolumab or camrelizumab (where available) are options for second-line treatment and beyond regardless of PD-L1 expression status. (See 'Efficacy in squamous cell cancer' below.)

The use of biomarkers other than PD-L1, TMB, or dMMR to select patients with adenocarcinoma for anti-PD-1 immunotherapy is in evolution. Pembrolizumab is no longer approved in the United States for treatment of refractory PD-L1-overexpressing esophageal, gastric, or EGJ adenocarcinomas. Yet, in Japan, nivolumab is approved for advanced gastric cancer that has progressed after conventional therapy, without regard to the number of prior regimens or specific biomarker expression. We would consider off-label use of pembrolizumab in patients with high levels of PD-L1 overexpression (ie, CPS ≥5) on a case by case basis, emphasizing the uncertainty of benefit, potential toxicity, and financial cost. (See 'Efficacy in adenocarcinoma' below.)

For eligible patients, we generally suggest a trial of immunotherapy for second line rather than a later line of therapy. (See 'Timing of immunotherapy' below.)

Approach to adenocarcinomas not eligible for immunotherapy

For most patients with adenocarcinoma who retain an excellent performance status and are not eligible for immunotherapy or human epidermal growth factor receptor 2 (HER2)-targeted therapy, we suggest ramucirumab plus paclitaxel after progression on or prior treatment with fluoropyrimidine- or platinum-containing chemotherapy. For patients with existing neuropathy who are sufficiently fit for a multiagent regimen, FOLFIRI plus ramucirumab is an alternative. (See 'Cytotoxic chemotherapy' below.)

The optimal second-line treatment strategy for patients with HER2-overexpressing, advanced gastroesophageal adenocarcinoma is unclear. Potential benefit has been shown for the antibody-drug conjugate fam-trastuzumab deruxtecan after progression on first-line trastuzumab, and this approach is reasonable, although myelosuppression and interstitial lung disease are notable toxicities. There has been no strong evidence to support continued targeting of HER2 beyond first-line trastuzumab, and this approach cannot be generally recommended. However, a weakness in most of the studies is that HER2 status was not reconfirmed prior to initiating the second-line HER2-directed therapy. If use of second-line trastuzumab is being considered, it is prudent to reconfirm HER2 positivity with repeat biopsy. (See 'HER2-positive disease and continued targeting of HER2 after progression' below.)

Cytotoxic chemotherapy with a regimen containing different agents than were used in the first-line regimen is another option. (See 'Cytotoxic chemotherapy' below.)

Trifluridine-tipiracil is a reasonable option for third-line therapy or beyond in patients who retain a good performance status after two or more other regimens. (See 'Trifluridine-tipiracil' below.)

Approach to squamous cell carcinoma ineligible for immunotherapy

For patients with squamous cell carcinoma (SCC) who retain an excellent performance status and who are not eligible for immunotherapy, utilization of other active cytotoxic chemotherapy agents not used in the first-line regimen is reasonable, either in combination or as serial single agents. For example, patients who received FOLFOX (oxaliplatin plus leucovorin and short-term infusional fluorouracil [FU]) initially could be offered single-agent irinotecan, a taxane, or FOLFIRI (irinotecan plus leucovorin and short-term infusional FU (table 1)). (See 'Cytotoxic chemotherapy' below.)

Not candidates for intensive therapy – For patients with either adenocarcinoma or SCC who are not eligible for anti-PD-1 immunotherapy and have only an adequate performance status, or those for whom limiting treatment-related toxicity is an important goal, we suggest monotherapy rather than combination chemotherapy. Options include irinotecan, weekly paclitaxel, weekly nanoparticle albumin-bound paclitaxel (nabpaclitaxel), apatinib (where available), or ramucirumab monotherapy (for patients with adenocarcinoma). (See 'Cytotoxic chemotherapy' below and 'Treatments targeting VEGF' below.)

EFFICACY OF INDIVIDUAL TREATMENTS

Cytotoxic chemotherapy — In general, clinical trials assessing the efficacy of a variety of second-line therapy regimens after failure of the first-line regimen in advanced esophagogastric cancer have shown that response rates are lower than they are in previously untreated patients, and toxicity rates tend to be higher [11-30].

The choice of regimen is empiric. No single regimen has emerged as a clear "winner," and few trials have compared different regimens [31,32]:

A Japanese trial comparing second-line irinotecan monotherapy (150 mg/m2 on days 1 and 15 every four weeks) with weekly paclitaxel (80 mg/m2 on days 1, 8, and 15 every four weeks) concluded that neither regimen was superior in terms of efficacy or tolerability [33]. A similar conclusion was reached in a second similar trial from South Korea [34].

No differences in outcomes could be shown in two trials comparing irinotecan plus cisplatin versus irinotecan alone, and in one trial comparing single-agent nanoparticle albumin-bound paclitaxel (nabpaclitaxel) versus solvent-based paclitaxel (although hypersensitivity reactions were less common with nabpaclitaxel) [10,35,36].

On the other hand, a randomized phase II trial of every three week docetaxel versus weekly paclitaxel in 78 patients with esophageal squamous cell carcinoma (SCC) refractory to fluoropyrimidine and platinum-based chemotherapy concluded that paclitaxel was more efficacious (response rate 26 versus 6 percent, median progression-free survival (PFS) 4.4 versus 2.1 months) and less toxic [37].

Combination versus monotherapy — Combination regimens containing agents not used in the first-line setting are also reasonable in the second line setting (eg, FOLFIRI [irinotecan plus leucovorin and short-term infusional fluorouracil (FU)], taxane-based therapy, or cisplatin plus FU after FOLFOX [oxaliplatin plus leucovorin and short-term infusional FU]). (See "Initial systemic therapy for locally advanced unresectable and metastatic esophageal and gastric cancer", section on 'Irinotecan-containing regimens' and "Initial systemic therapy for locally advanced unresectable and metastatic esophageal and gastric cancer", section on 'Docetaxel-containing'.)

However, monotherapy is an acceptable option. At least two small trials have failed to show a survival benefit but greater treatment-related toxicity of multiagent cytotoxic chemotherapy versus monotherapy in the second-line setting [9,10]. Quality of life and minimization of side effects are key considerations when choosing a cytotoxic chemotherapy regimen for second-line therapy, particularly for patients with a less than ideal performance status.

Any of these approaches could be considered appropriate for second-line treatment of adenocarcinomas or squamous cell cancers. For adenocarcinomas, there are no trials comparing any of these regimens with ramucirumab-containing therapy or trifluridine/tipiracil. (See 'Treatments restricted to adenocarcinomas' below.)

Checkpoint inhibitor immunotherapy — Immunotherapeutic approaches to cancer therapy are based on the premise that the immune system plays a key role in surveillance and eradication of malignancy and that tumors evolve ways to elude the immune system. We would only pursue immunotherapy if it was not administered in the frontline setting. We would generally not pursue anti-PD-1 immunotherapy for second-line therapy or beyond for patients who progressed while receiving prior PD-1 targeted therapy. There is no evidence to support benefit from a switch to a different immune checkpoint inhibitor for patients whose disease has progressed on one immune checkpoint inhibitor, regardless of histology. (See "Initial systemic therapy for locally advanced unresectable and metastatic esophageal and gastric cancer", section on 'Front-line immunotherapy'.)

Biomarkers and benefit from PD-1 inhibitors

Defective mismatch repair — For patients who have advanced SCC or adenocarcinoma of the esophagus or stomach with DNA mismatch repair deficiency (dMMR), we suggest pembrolizumab, if it was not administered for front-line therapy.

Tumors that lack the mismatch repair mechanism (ie, they have dMMR, the biologic footprint of which is high levels of microsatellite instability [MSI-H]) harbor many more mutations (ie, they are hypermutated) than do tumors of the same type without such mismatch repair defects. It is hypothesized that the neoantigens generated by mutations such as these are more immunogenic than those generated by other mutations.

Several steps are required for the immune system to effectively attack tumor cells. Several immune checkpoints exist to dampen the immune response in order to protect against detrimental inflammation and autoimmunity. In the setting of malignancy, such immune checkpoints can result in immune tolerance and contribute to unchecked tumor growth. Inhibition of these checkpoints might be expected to halt/reverse disease progression. One well-characterized checkpoint being targeted in several tumor types, including esophagogastric cancer, is programmed cell death-1 (PD-1). PD-1 is upregulated on activated T cells, and upon recognition of tumor via the T cell receptor, PD-1 engagement by PD-L1 expressed by the tumor or other immune cells infiltrating the tumor tissue can lead to T cell inactivation and a release of the "brake" on immune-mediated tumor eradication. (See "Principles of cancer immunotherapy".)

Proof of principle that cancers with dMMR might be particularly susceptible to inhibition of the PD-L1/PD-1 interaction was initially provided by a study of pembrolizumab in dMMR colorectal cancer. Subsequently, it has been established that immune checkpoint inhibitor immunotherapy appears to benefit a subset of patients with dMMR tumors, regardless of anatomic site of origin or tissue histology. As a result, in May 2017, the US Food and Drug Administration (FDA) approved pembrolizumab for treatment of a variety of advanced solid tumors, including gastric cancers, that had MSI-H or dMMR, that had progressed following prior treatment, and for which there were no satisfactory alternative treatment options, the first such approval of a tissue-agnostic anticancer treatment. (See "Tissue-agnostic cancer therapy: DNA mismatch repair deficiency, tumor mutational burden, and response to immune checkpoint blockade in solid tumors", section on 'Other tumors with MSI-H/dMMR'.)

Approximately 3 to 7 percent of esophagogastric adenocarcinomas are dMMR/MSI-H, most of which also overexpress PD-L1 [38-47]. Efficacy of pembrolizumab in dMMR esophagogastric cancer has been shown in the following studies:

In the phase II KEYNOTE-158 study, which enrolled 24 patients with gastric cancer [48], there were 11 objective responses (46 percent), four of which were complete, and median PFS was 11 months.

A subsequent post-hoc analysis of data from KEYNOTE-059 (third-line pembrolizumab), KEYNOTE-061 (second-line pembrolizumab versus chemotherapy) and KEYNOTE-062 (first-line pembrolizumab, versus pembrolizumab plus chemotherapy versus chemotherapy alone) also demonstrated that compared with the overall study population, the subset of patients with dMMR/MSI-H tumors experienced greater benefit from pembrolizumab alone or in combination with chemotherapy regardless of the line of treatment [47]. We would generally not pursue anti-PD-1 monotherapy for second-line therapy or beyond if disease progression occurred during prior PD-1-targeted therapy. (See "Initial systemic therapy for locally advanced unresectable and metastatic esophageal and gastric cancer", section on 'Front-line immunotherapy'.)

The approach to testing for dMMR is addressed in detail elsewhere. (See "Tissue-agnostic cancer therapy: DNA mismatch repair deficiency, tumor mutational burden, and response to immune checkpoint blockade in solid tumors", section on 'Assessing mismatch repair' and "Tissue-agnostic cancer therapy: DNA mismatch repair deficiency, tumor mutational burden, and response to immune checkpoint blockade in solid tumors", section on 'Approach to testing dMMR as a predictive marker'.)

High tumor mutational burden — Between 5 and 19 percent of gastric adenocarcinomas and approximately 2.4 percent of esophageal cancers have high levels of tumor mutational burden (TMB) [49,50], although this has been variably quantified. High TMB tumors have lower mutational levels than do those with dMMR, but TMB appears to be an independent biomarker of benefit for immune checkpoint inhibitor immunotherapy. Based upon an early report of the KEYNOTE-158 trial (which did not include patients with advanced esophageal or gastric cancer), pembrolizumab is now approved for patients with any solid tumor, including esophagogastric cancer, that has a TMB ≥10 mutations per megabase (mut/Mb), after progression on standard regimens. We would only pursue anti-PD-1 monotherapy for second-line therapy or beyond if prior PD-1 targeted therapy had not been administered front-line. (See "Tissue-agnostic cancer therapy: DNA mismatch repair deficiency, tumor mutational burden, and response to immune checkpoint blockade in solid tumors", section on 'Tumors with high mutational burden' and "Initial systemic therapy for locally advanced unresectable and metastatic esophageal and gastric cancer", section on 'Front-line immunotherapy'.)

Although it remains uncertain whether this is the appropriate threshold to define high TMB in esophagogastric cancer, particularly in patients without MSI [51], as validation studies were conducted mainly in lung and urothelial cancers, and thresholds for TMB are likely to vary across tumor types, we suggest using a next-generation sequencing (NGS) platform to assess TMB, and the use of a threshold TMB of ≥10 mut/Mb to select patients for immune checkpoint inhibitor immunotherapy. (See "Tissue-agnostic cancer therapy: DNA mismatch repair deficiency, tumor mutational burden, and response to immune checkpoint blockade in solid tumors", section on 'Approach to testing for high levels of TMB'.)

PD-L1 overexpression — The predictive value of programmed cell death ligand 1 (PD-L1) expression to select patients who might benefit from PD-1-blocking antibodies in esophagogastric cancer is in evolution [52]. PD-L1 expression is required for some indications but not others. In many cases, responses to PD-1 inhibitors seem to be less frequent (but not completely absent) in patients with low level or PD-L1-nonexpressing tumors. As examples (see 'Efficacy in adenocarcinoma' below and 'Efficacy in squamous cell cancer' below):

In the United States, pembrolizumab is approved as a second-line treatment for esophageal squamous cell cancers that express high levels of PD-L1 (combined positive score [CPS] ≥10) and we reserve this approach for those who did not receive first-line immunotherapy. (See 'Efficacy in squamous cell cancer' below.)

Pembrolizumab is no longer approved for third-line treatment of patients with esophagogastric adenocarcinomas and any level of PD-L1 expression. However, we would still consider off-label use on a case by case basis for those with adenocarcinoma and high levels of PD-1 expression (ie, CPS ≥10), as long as prior PD-1 targeted immunotherapy was not used front-line. (See 'Efficacy in adenocarcinoma' below.)

On the other hand, in the United States, nivolumab is approved for treatment of patients with unresectable, advanced, recurrent, or metastatic esophageal squamous cell cancer after prior fluoropyrimidine- and platinum-based chemotherapy, without regard for PD-L1 overexpression. (See 'Efficacy in squamous cell cancer' below.)

In Japan, nivolumab is approved for the treatment of unresectable, advanced, or recurrent gastric cancer that has progressed after "conventional chemotherapy," also without regard to specific biomarker expression.

Issues surrounding the assessment of PD-L1 overexpression are discussed in detail elsewhere. (See "Initial systemic therapy for locally advanced unresectable and metastatic esophageal and gastric cancer", section on 'PD-L1 expression status in upper gastrointestinal tract cancers'.)

Efficacy in adenocarcinoma

PembrolizumabPembrolizumab is an anti-PD-1 monoclonal antibody. Activity in advanced refractory PD-L1-expressing esophagogastric adenocarcinoma had been suggested in early phase I KEYNOTE-012 and 028 trials [53,54], and in the following later studies:

The superiority of pembrolizumab over systemic chemotherapy (investigators' choice of paclitaxel, docetaxel, or irinotecan) for second-line therapy in patients with an esophageal or esophagogastric junction (EGJ), Siewert type I (figure 1), cancer whose tumors express high levels of PD-L1 (CPS 10 or more) was shown in the phase III KEYNOTE-181 trial, in which the predominant population was SCC [55]. (See 'Efficacy in squamous cell cancer' below.)

In an early report of the KEYNOTE-059 cohort of 259 patients with previously treated gastric or EGJ adenocarcinoma, the objective response rate in the entire cohort was 11.6 percent (2.3 percent complete) [56]. Among those with PD-L1-positive tumors (CPS ≥1 percent), the objective response rate was 15.5 percent, with 13.5 percent partial and 2 percent complete responses. By contrast, the objective response rate in those with PD-L1-negative tumors was only 6.4 percent, with 2.8 percent experiencing a complete response. Grade 3 to 5 treatment-related adverse events developed in 18 percent, and two were fatal (acute kidney injury, pleural effusion).

Largely on the basis of these early data, pembrolizumab received accelerated approval from the FDA for treatment of patients with PD-L1-expressing recurrent or advanced gastric or EGJ adenocarcinoma who had received two or more lines of prior chemotherapy. The continued approval of this agent was contingent on positive data from the phase III second-line KEYNOTE-061 and first-line KEYNOTE-062 trials. Accelerated approval was withdrawn in January 2022 when these two trials failed to achieve their primary endpoints. However, given the potential for benefit, as shown in the KEYNOTE-181 and 061 trials, we would still consider off-label use on a case by case basis for those with adenocarcinoma and high levels of PD-1 expression (ie, CPS ≥10), as long as prior PD-1 targeted immunotherapy was not used front-line.

Data from these two trials are discussed below (See 'Timing of immunotherapy' below.)

Nivolumab – Early reports also suggest meaningful activity for nivolumab [57,58], another anti-PD-1 antibody, in patients with advanced, treatment-refractory gastroesophageal adenocarcinoma, not previously treated with PD-1 targeted immunotherapy:

The benefit of nivolumab was explored in a phase 3 trial (ONO-4538, the ATTRACTION-2 trial) in which 493 patients with advanced gastric or EGJ cancer who had failed two or more standard chemotherapy regimens were randomly assigned to nivolumab (3 mg/kg) or placebo every two weeks until unacceptable toxicity or disease progression [59]. The objective response rate with nivolumab was 11 percent, and median overall survival was modestly but significantly improved compared with placebo (5.3 versus 4.1 months [hazard ratio (HR) 0.63, 95% CI 0.51-0.78], 12-month survival 27 versus 11 percent); median PFS was also improved (1.61 versus 1.45 months [HR 0.60, 95% CI 0.49-0.75]). Severe (grade ≥3) adverse reactions were infrequent with nivolumab (11.5 versus 5.5 percent with placebo).

Notably, the trial did not restrict therapy to any subset of patients expressing a biomarker such as PD-L1 and it was conducted entirely in Asian populations.

A comparable degree of benefit in Western populations was suggested in the CheckMate-032 study, in which 160 patients with disease progression on or intolerance of at least one systemic chemotherapy regimen for advanced gastric, esophageal, or EGJ cancer were randomly assigned to nivolumab alone (3 mg/kg) every two weeks, nivolumab 1 mg/kg plus ipilimumab 3 mg/kg (nivo 1/ipi 3) every three weeks, or nivolumab 3 mg/kg plus ipilimumab 1 mg/kg (nivo 3/ipi 1) every three weeks [60]. Ipilimumab is another immune checkpoint inhibitor that targets a separate immune checkpoint, cytotoxic T-lymphocyte-associated protein 4.

Seventy-nine percent had received two or more prior therapies. Nivolumab, with or without ipilimumab, led to durable responses and long-term overall survival, and responses were observed regardless of tumor PD-L1 status. Twelve-month PFS rates for nivolumab alone, nivo 1/ipi 3, and nivo 3/ipi 1 were 8, 17, and 10 percent; the corresponding rates of 12-month survival were 39, 37, and 24 percent; and objective responses were seen in 12, 24, and 8 percent, respectively. Grade 3 or worse treatment-related adverse events were reported in 17, 47, and 27 percent of the three groups, respectively.

Given the lack of a chemotherapy control arm in these two trials, it is difficult to know if these results are better than could be achieved by cytotoxic chemotherapy.

Avelumab – On the other hand, benefit for avelumab, a PD-1 inhibitor, could not be shown in the phase III JAVELIN Gastric 300 trial, in which 371 patients failing two prior lines of systemic therapy were randomly assigned to avelumab or to systemic therapy of the clinician's choice (either weekly paclitaxel or irinotecan monotherapy; patients ineligible for systemic therapy received best supportive care only) [61]. Avelumab did not improve overall survival or PFS.

Efficacy in squamous cell cancer — The benefits of second-line therapies targeting the PD-1 pathway in esophageal SCC have been shown in the following trials:

Pembrolizumab – The superiority of pembrolizumab (an anti-PD-1 monoclonal antibody [MoAb]) over systemic chemotherapy (investigators' choice of paclitaxel, docetaxel, or irinotecan) for second-line therapy was shown in the phase III KEYNOTE-181 trial, which enrolled 628 patients with advanced/metastatic SCC (64 percent) or adenocarcinoma of the esophagus or EGJ, Siewert type I, unselected for PD-L1 expression status [55]. Among the 222 patients who had highly PD-L1-positive tumors (PD-L1 CPS ≥10, defined as the number of PD-L1-positive cells divided by the total number of tumor cells and then multiplied by 100), median overall survival was superior with pembrolizumab (9.3 versus 6.7 months), twice as many individuals remained alive at 12 months (43 versus 20 percent), and there were fewer grade 3 to 5 drug-related adverse events (18 versus 41 percent). The survival benefit in individuals with SCC (a coprimary endpoint) was also significant (median 8.2 versus 7.1 months, HR 0.78, 95% CI 0.63-0.96), but there was no improvement in overall survival with pembrolizumab in the entire intent-to-treat population, which included the 496 patients without high levels of PD-L1 overexpression (n = 628, median 7.1 months in both groups). In unplanned subgroup analysis, among those with CPS ≥10, the survival benefit was significant for SCC (HR 0.64, 95% CI 0.46-0.90) but not adenocarcinoma (HR 0.93, 95% CI 0.52-1.65). Additionally, when the analysis was limited to SCC, the survival benefit for pembrolizumab was restricted to those with CPS ≥10 and not lower levels.

Largely based on these data, in July 2019, the FDA approved single agent pembrolizumab for patients with recurrent locally advanced or metastatic SCC of the esophagus whose tumors express high levels of PD-L1 (CPS ≥10) and who have disease progression after one or more prior lines of systemic therapy (ie, second-line therapy).

NivolumabNivolumab, another anti-PD-1 MoAb, is also active in esophageal SCC, although it is not clear whether PD-L1 expression status is important to tumor response:

In an early, uncontrolled trial of nivolumab, objective responses were seen in 11 of 65 patients (17 percent) with refractory metastatic esophageal SCC, but the median time to progression was only 2.8 months; the safety profile was expected and manageable [57]. This trial did not restrict enrollment to any subset of patients expressing a biomarker such as PD-L1.

Support for second-line nivolumab was also provided by the phase 3 ATTRACTION-3 trial, in which 419 patients with previously treated (at least one fluoropyrimidine- and platinum-based regimen), advanced esophageal SCC were randomly assigned to nivolumab (240 mg every two weeks) or a single-agent taxane (investigators' choice of paclitaxel or docetaxel) [62]. Patients were unselected for biomarker expression; approximately one-half had PD-L1 expression levels in tumor cells <1 percent (using a different assay than that used to determine PD-L1 overexpression in the KEYNOTE-181 trial). Overall survival was significantly better with nivolumab (median 10.9 versus 8.4 months, HR for death 0.77, 95% CI 0.62-0.96), and many fewer patients had grade 3 or 4 treatment-related adverse effects (18 versus 63 percent). The survival benefit was independent of PD-L1 expression status in the tumor; survival benefit according to PD-L1 expression in immune and tumor cells (ie, the CPS) has not been reported, making it more difficult to compare directly with pembrolizumab. The survival advantage persisted with longer term follow-up, and nearly twice as many nivolumab-treated individuals were alive at three years (15.3 versus 8.7 percent) [63].

Largely based on the early report of this study, in June 2020, the FDA approved nivolumab for the treatment of patients with unresectable advanced, recurrent or metastatic esophageal SCC after prior fluoropyrimidine- and platinum-based chemotherapy, regardless of biomarker expression [64]. We would generally not pursue anti-PD-1 immunotherapy for second-line therapy or beyond for patients who progressed while receiving prior PD-1 targeted therapy. (See "Initial systemic therapy for locally advanced unresectable and metastatic esophageal and gastric cancer", section on 'Front-line immunotherapy'.)

Camrelizumab and tislelizumab

Camrelizumab, a third anti-PD-1 MoAb, which is approved in China for treatment of relapsed or refractory classical Hodgkin lymphoma [65], also appears to be active in advanced esophageal SCC [66]. In the randomized phase III ESCORT trial, in which 457 patients previously treated with first-line platinum-based chemotherapy were randomly assigned to camrelizumab or chemotherapy (docetaxel or irinotecan), second-line camrelizumab significantly improved overall survival (8.3 versus 6.2 months, HR 0.71, 95% CI 0.57-0.87), and benefit was irrespective of PD-L1 overexpression.

The anti-PD-1 monoclonal antibody tislelizumab is approved in China for a variety of indications, including advanced nasopharyngeal, urothelial, and non-small cell lung cancer. A significant survival benefit versus chemotherapy (irinotecan or taxane monotherapy) for second-line treatment of advanced or metastatic esophageal SCC was shown in the RATIONALE-302 trial (median 8.6 versus 6.3 months, HR 0.70, 95% CI 0.57-0.85), but the magnitude benefit was more pronounced in those with high levels of PD-L1 positivity (tumor area positivity ≥10 percent, median overall survival 10.3 versus 6.8 months) [67].

Timing of immunotherapy — For patients who are eligible for immunotherapy, we suggest PD-1 targeted immunotherapy rather than cytotoxic chemotherapy for second-line treatment after progression on the first-line regimen, as long as prior PD-1 targeted therapy was not administered first-line.

There is disagreement as to the appropriate timing (ie, second-line or beyond) for a trial of a PD-1 inhibitor in patients with advanced esophagogastric cancer that has not received frontline PD-1 targeted immunotherapy. In the United States, pembrolizumab is no longer approved for third-line treatment of PD-L1-overexpressing adenocarcinomas after failure of two separate chemotherapy regimens, but it is approved as a single agent for advanced or metastatic SCC of the esophagus that expresses PD-L1 at a CPS ≥10 and has progressed after one or more prior lines of systemic therapy, and for tumors with dMMR/MSI-H or high levels of TMB that have progressed following prior treatment and for which there are no satisfactory alternative treatment options. Nivolumab is approved for advanced SCC after prior fluoropyrimidine- and platinum-based chemotherapy, regardless of PD-L1 expression status.

Three trials have directly compared pembrolizumab or nivolumab versus cytotoxic chemotherapy in the second-line setting:

Pembrolizumab

In the KEYNOTE-061 trial, 592 patients with advanced gastric or EGJ cancer that had progressed on chemotherapy using a platinum and fluoropyrimidine combination were randomly assigned to pembrolizumab (200 mg every three weeks for up to two years) or paclitaxel monotherapy (80 mg/m2 on days 1, 8, and 15 every 28 days) [68]. Patients were initially enrolled without regard to PD-L1 overexpression, but enrollment was restricted to those with a CPS ≥1 after an interim analysis suggested inferior results with pembrolizumab in those with a CPS <1. When the analysis was limited to the 395 randomly allocated patients with a CPS ≥1, pembrolizumab did not significantly prolong overall survival (median 9.1 versus 8.3 months, HR 0.82, 95% CI 0.66-1.03), and the objective response rates were similar (16 versus 14 percent). However, the adverse event profile was better with pembrolizumab (grade 3 to 5 treatment-related adverse events in 14 versus 35 percent). The main differences were in fatigue, neuropathy, and cytopenia. In a post hoc analysis, which should be considered hypothesis-generating only, the pembrolizumab treatment effect appeared to be greater for patients with a PD-L1 CPS ≥10 and for patients whose tumors were MSI-H irrespective of the CPS.

This was confirmed in a later update of these results, presented at the 2020 American Society of Clinical Oncology (ASCO) annual meeting, in which the duration of response with pembrolizumab was significantly longer than with paclitaxel in the entire population of CPS ≥1 (median 19.1 versus 5.2 months), and in subgroup analysis, the overall survival benefit for pembrolizumab was greatest in those with PD-L1 CPS ≥10 [69]. The survival improvement with second-line pembrolizumab for the entire group with CPS ≥1 was 9.1 versus 8.3 months (HR 0.81, 95% CI 0.66-1.00), for those with CPS ≥5 it was 10.4 versus 8.3 months (HR 0.72, 95% CI 0.53-0.99), and for CPS ≥10 it was of a similar magnitude but not statistically significant (10.4 versus 8.0 months, HR 0.69, 95% CI 0.46-1.05).

The superiority of pembrolizumab over systemic chemotherapy (investigators' choice of paclitaxel, docetaxel, or irinotecan) for second-line therapy was also shown in the phase III KEYNOTE-181 trial, which enrolled 628 patients with advanced/metastatic SCC (63 percent of enrollees) or adenocarcinoma of the esophagus or EGJ, Siewert type I [55]. As noted above, among the 222 patients who had PD-L1-positive tumors (CPS ≥10), median overall survival was superior with pembrolizumab (9.3 versus 6.7 months), twice as many individuals remained alive at 12 months (43 versus 20 percent), and there were fewer grade 3 to 5 drug-related adverse events (18 versus 41 percent); neither survival nor health-related quality of life [70] were impacted by the choice of second-line therapy. In unplanned subset analysis, among those with CPS ≥10, a survival benefit favoring pembrolizumab was evident for SCC (HR 0.64, 95% CI 0.46-0.90) but not adenocarcinoma (HR 0.93, 95% CI 0.52-1.65). (See 'Efficacy in squamous cell cancer' above.)

Largely based on these data, in July 2019, the FDA approved pembrolizumab for patients with recurrent locally advanced or metastatic SCC of the esophagus whose tumors express high levels of PD-L1 (CPS ≥10) and who have disease progression after one or more prior lines of systemic therapy (ie, second-line therapy). (See 'Efficacy in squamous cell cancer' above.)

In our view, these data also provide support for off-label use of pembrolizumab in the second-line setting for individuals with highly PD-L1-overexpressing (CPS ≥10) adenocarcinomas of the esophagus, or EGJ, Siewert type I (figure 1).

Nivolumab Support for second-line nivolumab over chemotherapy was also provided by the phase 3 ATTRACTION-3 trial, in which 419 patients with previously treated (at least one fluoropyrimidine- and platinum-based regimen) advanced esophageal SCC were randomly assigned to nivolumab (240 mg every two weeks) or a single-agent taxane (investigators' choice of paclitaxel or docetaxel) [62]. Patients were unselected for biomarker expression. Overall survival was significantly better with nivolumab (median 10.9 versus 8.4 months, HR for death 0.77, 95% CI 0.62-0.96), and fewer patients had grade 3 or 4 treatment-related adverse effects. (See 'Efficacy in squamous cell cancer' above.)

Nivolumab is approved in the United States for treatment of advanced esophageal SCC after prior fluoropyrimidine and platinum-based chemotherapy, regardless of biomarker expression. In addition, in Japan, nivolumab is approved for the treatment of unresectable, advanced, or recurrent gastric cancer that has progressed after "conventional chemotherapy." We would generally not pursue anti-PD-1 monotherapy for second-line therapy or beyond if disease progression occurred during prior PD-1 targeted therapy.

Treatments restricted to adenocarcinomas

HER2-positive disease and continued targeting of HER2 after progression — The best treatment for HER2-positive adenocarcinomas after progression on a first-line trastuzumab-containing regimen is not established. To date, there has been no strong evidence to support continued targeting of HER2 with trastuzumab, lapatinib, or ado-trastuzumab emtansine, and we suggest not pursuing these approaches. A notable weakness in most of the studies evaluating second-line therapy targeting HER2 is that HER2 status was not reconfirmed prior to initiating the second-line HER2-directed therapy, and loss of HER2 expression could have contributed to the negative results of other HER2-targeted approaches. Some clinicians continue second-line trastuzumab in patients who are shown to retain HER2 positivity after progression on first-line trastuzumab, although this is controversial. Most recently, potential benefit has been shown for the antibody-drug conjugate fam-trastuzumab deruxtecan after progression on first-line trastuzumab, and this is a reasonable alternative approach for HER2-positive advanced adenocarcinoma, although treatment-related toxicity may be prominent.

Trastuzumab plus chemotherapy is a standard first-line treatment for human epidermal growth factor receptor 2 (HER2)-positive advanced esophagogastric cancer. (See "Initial systemic therapy for locally advanced unresectable and metastatic esophageal and gastric cancer", section on 'Benefit of trastuzumab'.)

Several anti-HER2 therapies have been examined in the second-line setting after failure of an initial trastuzumab-containing regimen:

Trastuzumab, lapatinib, or ado-trastuzumab emtansine – The benefit of continuing to target HER2 using trastuzumab, lapatinib, or ado-trastuzumab emtansine after progression on an initial trastuzumab-containing regimen has been addressed in the following trials, all of which failed to show benefit:

The benefit of continued trastuzumab after progression on an initial trastuzumab-containing regimen was studied in the phase II T-ACT trial in which 91 patients refractory to trastuzumab plus a fluoropyrimidine and platinum were randomly assigned to paclitaxel with or without trastuzumab [71]. There was no benefit from trastuzumab continued beyond progression. Notably, 69 percent of the patients whose tumor tissues were available lost tumoral HER2 positivity after progression on the prior trastuzumab-containing chemotherapy. No biomarker could be identified that was associated with efficacy of trastuzumab beyond progression.

The benefit of lapatinib, an inhibitor of both EGFR and HER2, in conjunction with weekly paclitaxel versus weekly paclitaxel for second-line therapy was evaluated in the Asian TyTAN trial, a two-phase, parallel-group phase III study that included an open-label dose optimization pilot study (n = 12) followed by a randomized trial (n = 261) in which patients with HER2-overexpressing gastric cancer were randomly assigned to paclitaxel (80 mg/m2 weekly) with or without lapatinib (1500 mg once daily) [72]. The addition of lapatinib to weekly paclitaxel provided no significant benefit in terms of overall survival (median 11 versus 8.9 months) or PFS (5.4 versus 4.4 months). However, the risk of death or disease progression was significantly lower in patients with IHC 3+ tumors who were treated with lapatinib, while it was not significantly different in those with IHC 0/1+ or IHC 2+ tumors. Importantly, few patients in either arm had received prior trastuzumab (7 percent of the lapatinib patients versus 15 percent of the placebo group). As a result, whether these results can be extrapolated to populations receiving initial trastuzumab is unclear.

A similar lack of benefit for second-line lapatinib was shown in a randomized phase II trial; of the 37 patients randomly assigned to lapatinib with or without capecitabine, 24 (65 percent) had received prior trastuzumab [73]. Only two patients (11 percent) achieved an objective partial response, both in the combined therapy group. The median time to progression was only 42 days with lapatinib monotherapy and 86 days with combined therapy.

There was also a lack of benefit for ado-trastuzumab emtansine (an antibody-drug conjugate composed of trastuzumab, a thioether linker, and a microtubule inhibitor) compared with a taxane alone for second-line therapy in the randomized phase II/III GATSBY trial [74].

One potential reason for the lack of efficacy of these second-line therapies targeting HER2 is loss of tumoral HER2 expression, which has been documented in 16 to 69 percent of such patients [71,75-77]. However, in at least one trial, no biomarker, including HER2 overexpression, could be identified that was associated with efficacy of trastuzumab beyond progression [71].

Fam-trastuzumab deruxtecan – Fam-trastuzumab deruxtecan is an antibody-drug conjugate composed of an anti-HER2 antibody, a cleavable tetrapeptide-based linker, and a cytotoxic topoisomerase I inhibitor; it is approved for locally advanced or metastatic HER2-positive gastric or EGJ adenocarcinoma who have received a prior trastuzumab-based regimen. (See "Systemic treatment for HER2-positive metastatic breast cancer", section on 'Fam-trastuzumab deruxtecan (preferred)'.)

In contrast to the above negative trials, benefit for fam-trastuzumab deruxtecan in previously treated HER2-positive gastric cancer was suggested in the open label randomized phase II DESTINY-Gastric01 trial, in which 187 patients with confirmed HER2-positive gastric cancer that had progressed while receiving two prior therapies, including trastuzumab, were randomly assigned to fam-trastuzumab deruxtecan or clinician's choice of chemotherapy (irinotecan [89 percent] or paclitaxel [11 percent]) [78]. HER2 status was assessed on the basis of the most recent archival tissue, and not systematically reconfirmed prior to enrollment. The fam-trastuzumab deruxtecan group had a significantly higher objective response rate (51 versus 14 percent), and significantly longer median overall survival (12.5 versus 8.4 months, HR for death 0.59, 95% CI 0.39-0.88) and median PFS (5.6 versus 3.5 months, HR 0.47, 95% CI 0.31-0.71). Objective response rates seemed higher in the subgroup with the highest level of HER2 expression (3+ or higher, 58 percent versus 29 percent in the control group), but the small numbers of patients in this subgroup precludes definitive conclusions.

Nausea, malaise, diarrhea, and vomiting occurred in over 25 percent of patients receiving fam-trastuzumab deruxtecan, but were mostly mild. Serious (grade 3 or 4) treatment-related toxicity included myelosuppression (neutropenia [51 percent], anemia [38 percent], thrombocytopenia [10 percent]), and interstitial lung disease (ILD; 10 percent); there was one treatment-related death classified as pneumonia. While there were no clinical adverse events of heart failure, 8 percent had asymptomatic grade 2 decrease in left ventricular ejection fraction (table 2).

Largely based on these data, the FDA approved fam-trastuzumab deruxtecan for treatment of adults with locally advanced, or metastatic HER2-overexpressing gastric or esophagogastric cancer junction adenocarcinoma who have received a prior trastuzumab-based regimen [79]. The recommended dose for gastric cancer is 6.4 mg/kg given as an IV infusion, once every three weeks until disease progression or unacceptable toxicity. The United States Prescribing Information for fam-trastuzumab deruxtecan recommends that clinicians monitor for and promptly investigate signs and symptoms including cough, dyspnea, fever, and other new or worsening respiratory symptoms, and that the drug be permanently discontinued for ≥grade 2 ILD/pneumonitis (table 3) or severe left ventricular dysfunction. Dose reduction guidelines are provided for severe myelosuppression.

Trifluridine-tipiracil — Efficacy for trifluridine-tipiracil was suggested in the TAGS trial, but the comparator arm was placebo and not an alternative chemotherapy regimen [80]. In this trial of 507 heavily pretreated patients (over 60 percent of the patients in each group had received three or more prior chemotherapy regimens) with adenocarcinoma of the stomach or EGJ, trifluridine-tipiracil (35 mg/m2 orally twice daily on days 1 through 5 and 8 through 12 of each 28-day cycle) significantly improved overall survival over placebo (median 5.7 versus 3.6 months) and was reasonably well tolerated. The most frequent grade 3 or higher adverse effects were neutropenia and anemia in the trifluridine-tipiracil group.

Largely based on these data, in February 2019, the FDA approved trifluridine-tipiracil for adult patients with metastatic gastric or gastroesophageal junction adenocarcinoma previously treated with at least two prior lines of chemotherapy that included a fluoropyrimidine, a platinum, either a taxane or irinotecan, and if appropriate, human epidermal growth factor receptor 2 (HER2)-targeted therapy.

An important point is that oral absorption of trifluridine-tipiracil appears to be adequate even in the setting of prior gastrectomy [81].

Treatments targeting VEGF — Elevated serum and tumor levels of vascular endothelial growth factor (VEGF) are associated with a poor prognosis in patients with resectable gastric adenocarcinoma [82-84]. In animal models of gastric adenocarcinoma, inhibition of vascular endothelial growth factor receptor (VEGFR)-2 reduces tumor growth and vascularity [85]. The importance of VEGFR-2 signaling as a therapeutic target in advanced gastric and EGJ adenocarcinomas was confirmed in the placebo-controlled phase III REGARD trial, which demonstrated a modest but significant survival benefit for the VEGFR-2 inhibitor ramucirumab after progression on first-line chemotherapy.

Ramucirumab with or without paclitaxel — We suggest ramucirumab plus paclitaxel rather than a different chemotherapy regimen for most patients with advanced or metastatic gastric or EGJ adenocarcinomas who have disease progression on or after prior treatment with fluoropyrimidine- or platinum-containing chemotherapy and retain an excellent performance status. For patients with an adequate performance status or those for whom limiting treatment-related toxicity is an important goal, ramucirumab monotherapy is an acceptable alternative.

Ramucirumab is a recombinant monoclonal antibody of the immunoglobulin G1 (IgG1) class that binds to VEGFR-2, blocking receptor activation. At least two trials have shown a survival benefit for therapy with ramucirumab, either as monotherapy (compared with best supportive care) or in combination with paclitaxel (over paclitaxel alone) in patients with previously treated, advanced gastric or EGJ adenocarcinoma [4,5]:

In the phase III REGARD trial, 355 patients with previously treated, advanced or metastatic gastric or EGJ adenocarcinoma were randomly assigned to best supportive care plus either ramucirumab (8 mg/kg intravenously every two weeks) or placebo [4]. Although the benefits were modest, patients treated with ramucirumab had significantly better median PFS (2.1 versus 1.3 months) and overall survival (5.2 versus 3.8 months, HR 0.78, 95% CI 0.60-0.99). The rate of objective tumor response was 8 versus 3 percent, but the overall disease control rate (objective response plus stable disease) was significantly higher with ramucirumab (49 versus 23 percent). The most common adverse event (all grades) in ramucirumab-treated patients was hypertension (16 versus 8 percent in the placebo group, 8 versus 3 percent grade 3 or worse). Ramucirumab was not associated with increased bleeding, venous thromboembolism, perforation, fistula formation, or proteinuria. There were three grade ≥3 arterial thromboembolism events in the ramucirumab arm versus none with placebo. (See "Toxicity of molecularly targeted antiangiogenic agents: Non-cardiovascular effects" and "Toxicity of molecularly targeted antiangiogenic agents: Cardiovascular effects".)

A survival benefit for ramucirumab was also demonstrated in the phase III RAINBOW trial, which compared weekly paclitaxel (80 mg/m2 on days 1, 8, and 15 of each 28-day cycle) plus either ramucirumab (8 mg/kg intravenously every two weeks) or placebo in 665 patients with metastatic gastric or EGJ adenocarcinoma who had disease progression on or within four months after first-line platinum- and fluoropyrimidine-based combination therapy [86]. Median overall survival was significantly better with ramucirumab (9.6 versus 7.4 months, HR 0.807, 95% CI 0.678-0.962), as was PFS (4.4 versus 2.9 months) and the objective response rate (28 versus 16 percent). Grade 3 or worse neutropenia was more common with ramucirumab (41 versus 19 percent), but rates of febrile neutropenia were low and similar in both groups (3 versus 2 percent). Rates of grade ≥3 hypertension were 14 versus 2 percent.

A similarly designed trial conducted in China (RAINBOW-Asia) in 440 patients with previously treated advanced gastric or EGJ adenocarcinoma concluded that combined therapy with ramucirumab plus weekly paclitaxel improved median PFS (4.14 versus 3.15 months) but not overall survival (median 8.71 versus 7.92 months, HR 0.963, 95% CI 0.771-1.203) compared with weekly paclitaxel alone, and rates of grade 3 or worse neutropenia (54 versus 39 percent) and febrile neutropenia (6 versus <1 percent) were worse [87].

Largely based on the REGARD trial results, the FDA approved single-agent ramucirumab for patients with advanced or metastatic gastric or EGJ cancer and disease progression during or after prior treatment with fluoropyrimidine- or platinum-containing chemotherapy. In November 2014, the combination of ramucirumab plus paclitaxel was also approved by the FDA for treatment of advanced gastric or EGJ cancer.

Whether ramucirumab plus paclitaxel is a preferred strategy over ramucirumab monotherapy is unknown given the lack of comparator trials. The REGARD and RAINBOW trials are not directly comparable in that patients in the REGARD trial were more heavily pretreated. In our view, ramucirumab plus weekly paclitaxel is a reasonable choice for patients who are willing to accept more treatment-related toxicity.

Ramucirumab plus FOLFIRI — Particularly for patients with pre-existing neuropathy, ramucirumab plus FOLFIRI is an alternative to ramucirumab plus paclitaxel.

There is some concern about the use of paclitaxel in patients who have received prior docetaxel in the first-line metastatic or neoadjuvant setting, given the potential for cross-resistance, and worsening of treatment-related neuropathy. (See "Adjuvant and neoadjuvant treatment of gastric cancer", section on 'FLOT'.)

The combination of ramucirumab plus irinotecan, leucovorin and short-term infusional FU (FOLFIRI) was directly compared with ramucirumab plus paclitaxel for second-line treatment in the randomized phase II RAMIRIS trial [88]. Although response rate were higher in the FOLFIRI group (22 versus 11 percent), overall survival was similar in both groups (median 6.8 versus 7.6 months, HR 0.97), as was treatment-related toxicity was similar. Among the 65 percent of patients who had previously received docetaxel, there was a higher objective response rate with FOLFIRI (25 versus 8 percent) but this did not translate into better overall survival (7.5 versus 6.6 months). Adverse events were similar in both groups, regardless of prior docetaxel exposure.

Other agents

Bevacizumab – The benefit of bevacizumab, a monoclonal antibody that binds to soluble VEGF and prevents binding to VEGFR, for gastric and EGJ adenocarcinomas is uncertain. Promising results were reported in a phase II study of bevacizumab in combination with cisplatin plus irinotecan in patients with gastric or EGJ adenocarcinoma [89]; however, a survival benefit for adding bevacizumab to capecitabine plus cisplatin could not be shown in the global phase III Avastin in Gastric Cancer (AVAGAST) trial [90]. A similar lack of benefit for the addition of bevacizumab to capecitabine plus cisplatin in Asian patients was also noted in a preliminary report of the AVATAR study presented at the 2012 ASCO Gastrointestinal Cancers Symposium [91].

Until further information is available, we suggest not using bevacizumab for patients with advanced esophagogastric cancer. Biomarkers may prove useful for identifying patients with advanced gastric adenocarcinoma who might benefit from the addition of bevacizumab to cytotoxic chemotherapy, but this is not yet established [92].

Apatinib – Apatinib (rivoceranib) is an orally active VEGFR-2 inhibitor that is approved for second-line treatment of advanced gastric cancer in China; it is not available in the United States or Europe. The data on activity in advanced gastric cancer are conflicting:

Activity was shown in a multicenter, randomized, double-blind trial in which 270 patients in China with advanced gastric cancer and prior failure on second-line chemotherapy were randomly assigned in a 2:1 ratio to apatinib (850 mg daily) or placebo [93]. Objective response rates were low with apatinib (3 versus 0 percent with placebo), but mean overall survival was modestly but significantly prolonged (median 6.5 versus 4.7 months).

On the other hand, activity for apatinib after failure of two prior chemotherapy regimens was not shown in a preliminary report of the placebo-controlled multinational phase III ANGEL trial [94]. The median overall survival was not significantly better with apatinib (5.78 versus 5.13 months, HR 0.93, p = 0.485).

RegorafenibRegorafenib is an orally active inhibitor of angiogenic (including VEGFR-1 to VEGFR-3), stromal, and oncogenic receptor tyrosine kinases. Activity was shown in a multicenter, randomized, double-blind phase II trial in which 152 patients with advanced gastric cancer and prior failure on first- or second-line chemotherapy were randomly assigned in a 2:1 ratio to regorafenib (160 mg daily) or placebo [95]. Median PFS was significantly longer with regorafenib (2.6 versus 0.9 months, HR 0.4, 95% CI 0.28-0.59), and there was a trend toward improved overall survival (median 5.8 versus 4.5 months, p = 0.47).

In our view, these data require confirmation before regorafenib can be considered a standard therapy for advanced gastric cancer.

Sunitinib and sorafenibSunitinib and sorafenib are small-molecule TKIs that inhibit VEGFR-1, VEGFR-2, and VEGFR-3, as well as other tyrosine kinases; they are referred to as "multitargeted" TKIs. Early studies have shown mixed results:

In a phase II trial of sunitinib monotherapy for second-line treatment of metastatic gastric adenocarcinoma, only 2 of 78 patients had an objective partial response (3 percent), while 25 others (32 percent) had stable disease [96]. Median overall survival was 6.8 months. Grade 3 or worse thrombocytopenia and neutropenia were reported in 35 and 30 percent, respectively.

The combination of sorafenib (400 mg orally, twice daily, continuously), docetaxel (75 mg/m2 intravenously on day 1), and cisplatin (75 mg/m2 intravenously on day 1) was studied in a phase II trial of 44 chemotherapy-naïve patients with locally advanced or metastatic (80 percent) gastric or EGJ adenocarcinoma [97]. Objective responses were noted in 41 percent, and median overall survival was 13.6 months; the major grade 3 or 4 toxicity was neutropenia (64 percent).

However, benefit for sunitinib could not be confirmed in the only randomized trial, in which sunitinib (37.5 mg every day) plus docetaxel (60 mg/m2 every three weeks) was compared with docetaxel monotherapy (60 mg/m2 every three weeks) for second-line treatment of patients with unresectable or metastatic gastric cancer (n = 107) [98]. Although combined therapy was associated with a significantly higher objective response rate, neither time to disease progression (the primary endpoint, 3.9 versus 2.6 months) nor overall survival (8.0 versus 6.6 months) was significantly improved.

Aflibercept – Intravenous aflibercept, a recombinant fusion protein consisting of the VEGF-binding portions from human VEGFR-1 and VEGFR-2 fused to the fragment crystallizable (Fc) portion of human IgG1, functions as a decoy receptor that prevents intravascular and extravascular VEGF-A, VEGF-B, and placental growth factor (PlGF) from binding to their receptors. Unfortunately, benefit from the addition of aflibercept to FOLFOX could not be shown in a randomized phase II trial [99].

SPECIAL CONSIDERATIONS DURING THE COVID-19 PANDEMIC — The COVID-19 pandemic has increased the complexity of cancer care. Important issues in areas where viral transmission rates are high include balancing the risk from delaying diagnostic evaluation and cancer treatment versus harm from COVID-19, minimizing the number of clinic and hospital visits to reduce exposure whenever possible, mitigating the negative impacts of social distancing on delivery of care, and appropriately and fairly allocating limited health care resources. Recommendations for vaccination of cancer patients and cancer care during active phases of the COVID-19 pandemic are discussed separately. (See "COVID-19: Considerations in patients with cancer".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Gastric cancer" and "Society guideline links: Esophageal cancer".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: Esophageal cancer (The Basics)" and "Patient education: Stomach cancer (The Basics)")

SUMMARY AND RECOMMENDATIONS

Goals of therapy, patient selection, and regimen choice

The major goals of systemic chemotherapy for advanced esophagogastric cancer are symptom palliation, improved quality of life, and prolonged survival. (See 'Approach to the patient' above.)

Decision making for second line treatment must take into account performance status, comorbidity, goals of care, patient preference, the overall medical condition, and disease status.

-For patients with a poor performance status, poorly controlled comorbidity, or a preference for no additional therapy, we suggest supportive care alone. (See 'Patient selection' above.)

-For patients who retain an adequate performance status, preference, symptom burden, quality of life, histologic tumor type, genomic testing, and the nature of the first-line regimen are key considerations. Somatic and germline genomic testing is indicated for those who might be eligible for immunotherapy or molecularly targeted therapy.

For patients who received prior programmed cell death 1 (PD-1) targeted therapy, we would generally not pursue rechallenge unless prior anti-PD-1 immunotherapy was discontinued due to a specific toxicity that has since resolved, and possibly for patients with progression at few sites after a prior objective response or prolonged period of stable disease. (See 'Regimen choice' above.)

Our approach – If trials are unavailable or patients are ineligible, our general approach is summarized below:

Deficient mismatch repair or high TMB – For patients who have either DNA mismatch repair deficiency or high levels of tumor mutational burden (TMB), we suggest pembrolizumab rather than cytotoxic chemotherapy, if it was not administered for frontline therapy (Grade 2C). (See 'Defective mismatch repair' above and 'High tumor mutational burden' above.)

Other adenocarcinomas

-HER2-positive – The optimal second-line treatment strategy for patients with HER2-overexpressing advanced gastroesophageal adenocarcinoma is not established. A trial of fam-trastuzumab deruxtecan after progression on first-line trastuzumab is reasonable, although myelosuppression and interstitial lung disease are notable toxicities. For most patients, we suggest not pursuing second-line trastuzumab, lapatinib, or ado-trastuzumab emtansine (Grade 2B). (See 'HER2-positive disease and continued targeting of HER2 after progression' above.)

Another alternative is cytotoxic chemotherapy.

-HER2-negative, PD-L1 expression – An option for treatment at progression for patients who have high levels of programmed cell death ligand 1 (PD-L1) expression (combined positive score [CPS] ≥10) is off-label use of pembrolizumab (table 4), if an immune checkpoint inhibitor was not administered for front-line therapy. (See 'Efficacy in adenocarcinoma' above.)

Another alternative is cytotoxic chemotherapy

-Cytotoxic chemotherapy – For most patients with HER2-negative non-PD-1 overexpressing adenocarcinomas who have disease progression with fluoropyrimidine- and platinum-containing chemotherapy, retain an excellent performance status, a favorable comorbidity profile, and a preference for intensive treatment we suggest ramucirumab plus paclitaxel rather than a different regimen (Grade 2C). For patients with pre-existing neuropathy, ramucirumab plus FOLFIRI is an alternative. (See 'Ramucirumab with or without paclitaxel' above and 'Ramucirumab plus FOLFIRI' above.)

For patients with an adequate performance status and comorbidity profile or those for whom limiting treatment-related toxicity is an important goal, we suggest monotherapy rather than combination chemotherapy (Grade 2C). Options include irinotecan, weekly paclitaxel, apatinib (where available), or ramucirumab monotherapy. (See 'Combination versus monotherapy' above and 'Treatments targeting VEGF' above.)

Trifluridine-tipiracil is a reasonable option for third-line therapy or beyond for patients who retain an adequate performance status. (See 'Trifluridine-tipiracil' above.)

Squamous cell cancer

-Second-line pembrolizumab is an option for those with high levels of PD-L1 expression (CPS ≥10), and nivolumab or camrelizumab (where available) are options for second-line treatment and beyond regardless of PD-L1 expression status, if an immune checkpoint inhibitor was not administered for front-line therapy. (See 'Efficacy in squamous cell cancer' above.)

-For patients with who are not eligible for immunotherapy and who retain an excellent performance status, a favorable comorbidity profile and preference for intensive treatment, we suggest a combination regimen containing agents not used in the first-line setting (Grade 2C). Options for patients treated with initial FOLFOX include FOLFIRI (irinotecan plus leucovorin and short-term infusional fluorouracil [FU], (table 1)), taxane-based therapy, or cisplatin plus FU. (See 'Cytotoxic chemotherapy' above.)

-For patients with an adequate performance status and comorbidity profile or those for whom limiting treatment-related toxicity is an important goal, we suggest monotherapy rather than combination chemotherapy (Grade 2C). Options include irinotecan, weekly paclitaxel, or weekly nabpaclitaxel. (See 'Cytotoxic chemotherapy' above and 'Treatments targeting VEGF' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Johanna Bendell, MD, who contributed to an earlier version of this topic review.

  1. Kang JH, Lee SI, Lim DH, et al. Salvage chemotherapy for pretreated gastric cancer: a randomized phase III trial comparing chemotherapy plus best supportive care with best supportive care alone. J Clin Oncol 2012; 30:1513.
  2. 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 2011; 47:2306.
  3. Ford HE, 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 2014; 15:78.
  4. 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 2014; 383:31.
  5. Wilke H, Van Cutsem E, Oh SC, et al. RAINBOW: A global, phase III, randomized, double-blind study of ramucirumab plus paclitaxel versus placebo plus paclitaxel in the treatment of metastatic gastroesophageal junction (GEJ) and gastric adenocarcinoma following disease progression on first-line platinum- and fluoropyrimidine-containing combination therapy rainbow IMCL CP12-0922 (I4T-IE-JVBE). J Clin Oncol 2014; 32S: ASCO #LBA7.
  6. Tomita Y, Moldovan M, Chang Lee R, et al. Salvage systemic therapy for advanced gastric and oesophago-gastric junction adenocarcinoma. Cochrane Database Syst Rev 2020; 11:CD012078.
  7. Catalano V, Graziano F, Santini D, et al. Second-line chemotherapy for patients with advanced gastric cancer: who may benefit? Br J Cancer 2008; 99:1402.
  8. Chakravarty D, Johnson A, Sklar J, et al. Somatic Genomic Testing in Patients With Metastatic or Advanced Cancer: ASCO Provisional Clinical Opinion. J Clin Oncol 2022; 40:1231.
  9. Sym SJ, Hong J, Park J, et al. A randomized phase II study of biweekly irinotecan monotherapy or a combination of irinotecan plus 5-fluorouracil/leucovorin (mFOLFIRI) in patients with metastatic gastric adenocarcinoma refractory to or progressive after first-line chemotherapy. Cancer Chemother Pharmacol 2013; 71:481.
  10. Higuchi K, Tanabe S, Shimada K, et al. Biweekly irinotecan plus cisplatin versus irinotecan alone as second-line treatment for advanced gastric cancer: a randomised phase III trial (TCOG GI-0801/BIRIP trial). Eur J Cancer 2014; 50:1437.
  11. Lorenzen S, Duyster J, Lersch C, et al. Capecitabine plus docetaxel every 3 weeks in first- and second-line metastatic oesophageal cancer: final results of a phase II trial. Br J Cancer 2005; 92:2129.
  12. Gold PJ, Goldman B, Iqbal S, et al. Cetuximab as second-line therapy in patients with metastatic esophageal cancer: a phase II Southwest Oncology Group Study. J Clin Oncol 2008; 26S: ASCO #4536.
  13. Kim Y, Park S, Park J, et al. Oxaliplatin, 5-fluorouracil, and leuvocorin and (FOLFOX-4) combination chemotherapy as salvage treatment in pretreated patients with advanced gastric cancer (AGC). J Clin Oncol 2008; 26S: ASCO #4577.
  14. Kodera Y, Ito S, Mochizuki Y, et al. A phase II study of weekly paclitaxel as second-line chemotherapy for advanced gastric Cancer (CCOG0302 study). Anticancer Res 2007; 27:2667.
  15. Hartmann JT, Pintoffl JP, Al-Batran SE, et al. Mitomycin C plus infusional 5-fluorouracil in platinum-refractory gastric adenocarcinoma: an extended multicenter phase II study. Onkologie 2007; 30:235.
  16. Yoshida T, Yoshikawa T, Tsuburaya A, et al. Feasibility study of biweekly CPT-11 plus CDDP for S-1- and paclitaxel-refractory, metastatic gastric cancer. Anticancer Res 2006; 26:1595.
  17. Cho BC, Kim JH, Kim CB, et al. Paclitaxel and leucovorin-modulated infusional 5-fluorouracil combination chemotherapy for metastatic gastric cancer. Oncol Rep 2006; 15:621.
  18. Sym SJ, Ryu MH, Lee JL, et al. Salvage chemotherapy with biweekly irinotecan, plus 5-fluorouracil and leucovorin in patients with advanced gastric cancer previously treated with fluoropyrimidine, platinum, and taxane. Am J Clin Oncol 2008; 31:151.
  19. Chun JH, Kim HK, Lee JS, et al. Weekly irinotecan in patients with metastatic gastric cancer failing cisplatin-based chemotherapy. Jpn J Clin Oncol 2004; 34:8.
  20. Kim SH, Lee GW, Go SI, et al. A phase II study of irinotecan, continuous 5-fluorouracil, and leucovorin (FOLFIRI) combination chemotherapy for patients with recurrent or metastatic gastric cancer previously treated with a fluoropyrimidine-based regimen. Am J Clin Oncol 2010; 33:572.
  21. Giuliani F, Gebbia V, De Vita F, et al. Docetaxel as salvage therapy in advanced gastric cancer: a phase II study of the Gruppo Oncologico Italia Meridionale (G.O.I.M.). Anticancer Res 2003; 23:4219.
  22. Cascinu S, Graziano F, Cardarelli N, et al. Phase II study of paclitaxel in pretreated advanced gastric cancer. Anticancer Drugs 1998; 9:307.
  23. Burkart C, Bokemeyer C, Klump B, et al. A phase II trial of weekly irinotecan in cisplatin-refractory esophageal cancer. Anticancer Res 2007; 27:2845.
  24. Lordick F, von Schilling C, Bernhard H, et al. Phase II trial of irinotecan plus docetaxel in cisplatin-pretreated relapsed or refractory oesophageal cancer. Br J Cancer 2003; 89:630.
  25. Hironaka S, Zenda S, Boku N, et al. Weekly paclitaxel as second-line chemotherapy for advanced or recurrent gastric cancer. Gastric Cancer 2006; 9:14.
  26. Graziano F, Catalano V, Baldelli AM, et al. A phase II study of weekly docetaxel as salvage chemotherapy for advanced gastric cancer. Ann Oncol 2000; 11:1263.
  27. Seo HY, Kim DS, Choi YS, et al. Treatment outcomes of oxaliplatin, 5-FU, and leucovorin as salvage therapy for patients with advanced or metastatic gastric cancer: a retrospective analysis. Cancer Chemother Pharmacol 2009; 63:433.
  28. Jeong J, Jeung HC, Rha SY, et al. Phase II study of combination chemotherapy of 5-fluorouracil, low-dose leucovorin, and oxaliplatin (FLOX regimen) in pretreated advanced gastric cancer. Ann Oncol 2008; 19:1135.
  29. Baize N, Abakar-Mahamat A, Mounier N, et al. Phase II study of paclitaxel combined with capecitabine as second-line treatment for advanced gastric carcinoma after failure of cisplatin-based regimens. Cancer Chemother Pharmacol 2009; 64:549.
  30. Lee JL, Ryu MH, Chang HM, et al. A phase II study of docetaxel as salvage chemotherapy in advanced gastric cancer after failure of fluoropyrimidine and platinum combination chemotherapy. Cancer Chemother Pharmacol 2008; 61:631.
  31. Wesolowski R, Lee C, Kim R. Is there a role for second-line chemotherapy in advanced gastric cancer? Lancet Oncol 2009; 10:903.
  32. Thallinger CM, Raderer M, Hejna M. Esophageal cancer: a critical evaluation of systemic second-line therapy. J Clin Oncol 2011; 29:4709.
  33. Hironaka S, Ueda S, Yasui H, et al. Randomized, open-label, phase III study comparing irinotecan with paclitaxel in patients with advanced gastric cancer without severe peritoneal metastasis after failure of prior combination chemotherapy using fluoropyrimidine plus platinum: WJOG 4007 trial. J Clin Oncol 2013; 31:4438.
  34. Lee KW, Maeng CH, Kim TY, et al. A Phase III Study to Compare the Efficacy and Safety of Paclitaxel Versus Irinotecan in Patients with Metastatic or Recurrent Gastric Cancer Who Failed in First-line Therapy (KCSG ST10-01). Oncologist 2019; 24:18.
  35. Nishikawa K, Fujitani K, Inagaki H, et al. Randomised phase III trial of second-line irinotecan plus cisplatin versus irinotecan alone in patients with advanced gastric cancer refractory to S-1 monotherapy: TRICS trial. Eur J Cancer 2015; 51:808.
  36. Shitara K, Takashima A, Fujitani K, et al. Nab-paclitaxel versus solvent-based paclitaxel in patients with previously treated advanced gastric cancer (ABSOLUTE): an open-label, randomised, non-inferiority, phase 3 trial. Lancet Gastroenterol Hepatol 2017; 2:277.
  37. Yamamoto S, Kawakami H, Kii T, et al. Randomized phase II study of docetaxel versus paclitaxel in patients with esophageal squamous cell carcinoma refractory to fluoropyrimidine- and platinum-based chemotherapy: OGSG1201. Eur J Cancer 2021; 154:307.
  38. Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017; 357:409.
  39. An JY, Kim H, Cheong JH, et al. Microsatellite instability in sporadic gastric cancer: its prognostic role and guidance for 5-FU based chemotherapy after R0 resection. Int J Cancer 2012; 131:505.
  40. Kim H, An JY, Noh SH, et al. High microsatellite instability predicts good prognosis in intestinal-type gastric cancers. J Gastroenterol Hepatol 2011; 26:585.
  41. Beghelli S, de Manzoni G, Barbi S, et al. Microsatellite instability in gastric cancer is associated with better prognosis in only stage II cancers. Surgery 2006; 139:347.
  42. Mathiak M, Warneke VS, Behrens HM, et al. Clinicopathologic Characteristics of Microsatellite Instable Gastric Carcinomas Revisited: Urgent Need for Standardization. Appl Immunohistochem Mol Morphol 2017; 25:12.
  43. Pietrantonio F, Miceli R, Raimondi A, et al. Individual Patient Data Meta-Analysis of the Value of Microsatellite Instability As a Biomarker in Gastric Cancer. J Clin Oncol 2019; 37:3392.
  44. Farris AB 3rd, Demicco EG, Le LP, et al. Clinicopathologic and molecular profiles of microsatellite unstable Barrett Esophagus-associated adenocarcinoma. Am J Surg Pathol 2011; 35:647.
  45. Falkenback D, Johansson J, Halvarsson B, Nilbert M. Defective mismatch-repair as a minor tumorigenic pathway in Barrett esophagus-associated adenocarcinoma. Cancer Genet Cytogenet 2005; 157:82.
  46. Middha S, Zhang L, Nafa K, et al. Reliable Pan-Cancer Microsatellite Instability Assessment by Using Targeted Next-Generation Sequencing Data. JCO Precis Oncol 2017; 2017.
  47. Chao J, Fuchs CS, Shitara K, et al. Assessment of Pembrolizumab Therapy for the Treatment of Microsatellite Instability-High Gastric or Gastroesophageal Junction Cancer Among Patients in the KEYNOTE-059, KEYNOTE-061, and KEYNOTE-062 Clinical Trials. JAMA Oncol 2021; 7:895.
  48. Marabelle A, Le DT, Ascierto PA, et al. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J Clin Oncol 2020; 38:1.
  49. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med 2017; 9:34.
  50. Parikh AR, He Y, Hong TS, et al. Analysis of DNA Damage Response Gene Alterations and Tumor Mutational Burden Across 17,486 Tubular Gastrointestinal Carcinomas: Implications for Therapy. Oncologist 2019; 24:1340.
  51. Foote MB, Maron SB, Cercek A, et al. TMB cut-offs fail to predict benefit of PD-1 blockade in gastroesophageal adenocarcinoma in KEYNOTE-061. Ann Oncol 2021; 32:1188.
  52. Lu S, Stein JE, Rimm DL, et al. Comparison of Biomarker Modalities for Predicting Response to PD-1/PD-L1 Checkpoint Blockade: A Systematic Review and Meta-analysis. JAMA Oncol 2019; 5:1195.
  53. Muro K, Chung HC, Shankaran V, et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1b trial. Lancet Oncol 2016; 17:717.
  54. Doi T, Piha-Paul SA, Jalal SI, et al. Safety and Antitumor Activity of the Anti-Programmed Death-1 Antibody Pembrolizumab in Patients With Advanced Esophageal Carcinoma. J Clin Oncol 2018; 36:61.
  55. 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 2020; 38:4138.
  56. 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 2018; 4:e180013.
  57. Kudo T, Hamamoto Y, Kato K, et al. Nivolumab treatment for oesophageal squamous-cell carcinoma: an open-label, multicentre, phase 2 trial. Lancet Oncol 2017; 18:631.
  58. Kang YK, Satoh T, Ryu MH, et al. Nivolumab (ONO-4538/BMS-936558) as salvage treatment after second or later-line chemotherapy for advanced gastric or gastro-esophageal junction cancer (AGC): A double-blinded, randomized, phase III trial. J Clin Oncol 2017; 35S: ASCO #2.
  59. Kang YK, Boku N, Satoh T, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017; 390:2461.
  60. Janjigian YY, Bendell J, Calvo E, et al. CheckMate-032 Study: Efficacy and Safety of Nivolumab and Nivolumab Plus Ipilimumab in Patients With Metastatic Esophagogastric Cancer. J Clin Oncol 2018; 36:2836.
  61. Bang YJ, Ruiz EY, Van Cutsem E, et al. Phase III, randomised trial of avelumab versus physician's choice of chemotherapy as third-line treatment of patients with advanced gastric or gastro-oesophageal junction cancer: primary analysis of JAVELIN Gastric 300. Ann Oncol 2018; 29:2052.
  62. Kato K, Cho BC, Takahashi M, et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019; 20:1506.
  63. Okada M, Kato K, Cho BC, et al. Three-Year Follow-Up and Response-Survival Relationship of Nivolumab in Previously Treated Patients with Advanced Esophageal Squamous Cell Carcinoma (ATTRACTION-3). Clin Cancer Res 2022; 28:3277.
  64. Nivolumab injection. United States Prescribing Information. US National Library of Medicine. http://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125554s081lbl.pdf (Accessed on June 11, 2020).
  65. Markham A, Keam SJ. Camrelizumab: First Global Approval. Drugs 2019; 79:1355.
  66. Huang J, Xu J, Chen Y, et al. Camrelizumab versus investigator's choice of chemotherapy as second-line therapy for advanced or metastatic oesophageal squamous cell carcinoma (ESCORT): a multicentre, randomised, open-label, phase 3 study. Lancet Oncol 2020; 21:832.
  67. Shen L, Kato K, Kim SB, et al. Tislelizumab Versus Chemotherapy as Second-Line Treatment for Advanced or Metastatic Esophageal Squamous Cell Carcinoma (RATIONALE-302): A Randomized Phase III Study. J Clin Oncol 2022; 40:3065.
  68. Shitara K, Özgüroğlu M, Bang YJ, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet 2018; 392:123.
  69. Fuchs CS, Özgüroğlu M, Bagn YJ, et al. Pembrolizumab versus paclitaxel for previously treated patients with PD-L1–positive advanced gastric or gastroesophageal junction cancer (GC): Update from the phase III KEYNOTE-061 trial. J Clin Oncol 38:2020 )suppl; abstr 4503). Abstract available online at: https://meetinglibrary.asco.org/record/186716/abstract (Accessed on February 09, 2022).
  70. Adenis A, Kulkarni AS, Girotto GC, et al. Impact of Pembrolizumab Versus Chemotherapy as Second-Line Therapy for Advanced Esophageal Cancer on Health-Related Quality of Life in KEYNOTE-181. J Clin Oncol 2022; 40:382.
  71. Makiyama A, Sukawa Y, Kashiwada T, et al. Randomized, Phase II Study of Trastuzumab Beyond Progression in Patients With HER2-Positive Advanced Gastric or Gastroesophageal Junction Cancer: WJOG7112G (T-ACT Study). J Clin Oncol 2020; 38:1919.
  72. Satoh T, Xu RH, Chung HC, et al. Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN--a randomized, phase III study. J Clin Oncol 2014; 32:2039.
  73. Lorenzen S, Riera Knorrenschild J, Haag GM, et al. Lapatinib versus lapatinib plus capecitabine as second-line treatment in human epidermal growth factor receptor 2-amplified metastatic gastro-oesophageal cancer: a randomised phase II trial of the Arbeitsgemeinschaft Internistische Onkologie. Eur J Cancer 2015; 51:569.
  74. Thuss-Patience PC, Shah MA, Ohtsu A, et al. Trastuzumab emtansine versus taxane use for previously treated HER2-positive locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma (GATSBY): an international randomised, open-label, adaptive, phase 2/3 study. Lancet Oncol 2017; 18:640.
  75. Saeki H, Oki E, Kashiwada T, et al. Re-evaluation of HER2 status in patients with HER2-positive advanced or recurrent gastric cancer refractory to trastuzumab (KSCC1604). Eur J Cancer 2018; 105:41.
  76. Janjigian YY, Sanchez-Vega F, Jonsson P, et al. Genetic Predictors of Response to Systemic Therapy in Esophagogastric Cancer. Cancer Discov 2018; 8:49.
  77. Pietrantonio F, Caporale M, Morano F, et al. HER2 loss in HER2-positive gastric or gastroesophageal cancer after trastuzumab therapy: Implication for further clinical research. Int J Cancer 2016; 139:2859.
  78. Shitara K, Bang YJ, Iwasa S, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N Engl J Med 2020; 382:2419.
  79. Fam-trastuzumab deruxtecan-nxki injection. United States Prescribing Information. US National Library of Medicine. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/761139s011lbl.pdf (Accessed on January 19, 2021).
  80. Shitara K, Doi T, Dvorkin M, et al. Trifluridine/tipiracil versus placebo in patients with heavily pretreated metastatic gastric cancer (TAGS): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2018; 19:1437.
  81. Ilson DH, Tabernero J, Prokharau A, et al. Efficacy and Safety of Trifluridine/Tipiracil Treatment in Patients With Metastatic Gastric Cancer Who Had Undergone Gastrectomy: Subgroup Analyses of a Randomized Clinical Trial. JAMA Oncol 2020; 6:e193531.
  82. Pera M, Vidal O, Elizade J, et al. Tumor VEGF expression and serum VEGF levels are independent prognostic factors in curatively resected gastric cancer patients. J Clin Oncol 2006; 24S: ASCO #4021.
  83. Fondevila C, Metges JP, Fuster J, et al. p53 and VEGF expression are independent predictors of tumour recurrence and survival following curative resection of gastric cancer. Br J Cancer 2004; 90:206.
  84. Yao JC, Wang L, Wei D, et al. Association between expression of transcription factor Sp1 and increased vascular endothelial growth factor expression, advanced stage, and poor survival in patients with resected gastric cancer. Clin Cancer Res 2004; 10:4109.
  85. Jung YD, Mansfield PF, Akagi M, et al. Effects of combination anti-vascular endothelial growth factor receptor and anti-epidermal growth factor receptor therapies on the growth of gastric cancer in a nude mouse model. Eur J Cancer 2002; 38:1133.
  86. 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 2014; 15:1224.
  87. Xu RH, Zhang Y, Pan H, et al. Efficacy and safety of weekly paclitaxel with or without ramucirumab as second-line therapy for the treatment of advanced gastric or gastroesophageal junction adenocarcinoma (RAINBOW-Asia): a randomised, multicentre, double-blind, phase 3 trial. Lancet Gastroenterol Hepatol 2021; 6:1015.
  88. Lorenzen S, Thuss-Patience P, Pauligk C, et al. FOLFIRI plus ramucirumab versus paclitaxel plus ramucirumab as second-line therapy for patients with advanced or metastatic gastroesophageal adenocarcinoma with or without prior docetaxel - results from the phase II RAMIRIS Study of the German Gastric Cancer Study Group at AIO. Eur J Cancer 2022; 165:48.
  89. Shah MA, Ramanathan RK, Ilson DH, et al. Multicenter phase II study of irinotecan, cisplatin, and bevacizumab in patients with metastatic gastric or gastroesophageal junction adenocarcinoma. J Clin Oncol 2006; 24:5201.
  90. Ohtsu A, Shah MA, Van Cutsem E, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol 2011; 29:3968.
  91. Shen L, Li J, XU Jm, et al. Efficacy and tolerability of bevacizumab (BEV) plus capecitabine and cisplatin (XP) in Chinese patients (pts) with locally advanced or metastatic gastric/gastroesophageal junction cancer (AGC): Results from the AVATAR study. J Clin Oncol 2012; 30S: ASCO #73.
  92. Van Cutsem E, de Haas S, Kang YK, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a biomarker evaluation from the AVAGAST randomized phase III trial. J Clin Oncol 2012; 30:2119.
  93. Li J, Qin S, Xu J, et al. Randomized, Double-Blind, Placebo-Controlled Phase III Trial of Apatinib in Patients With Chemotherapy-Refractory Advanced or Metastatic Adenocarcinoma of the Stomach or Gastroesophageal Junction. J Clin Oncol 2016; 34:1448.
  94. Kang YK, Kang WK, Di Bartolomeo M, et al. Randomized phase III ANGEL study of rivoceranib (apatinib) + best supportive care (BSC) vs placebo + BSC in patients with advanced/metastatic gastric cancer who failed ≥2 prior chemotherapy regimens. Ann Oncol 2019; 30S: ESMO #LBA43.
  95. Pavlakis N, Sjoquist KM, Martin AJ, et al. Regorafenib for the Treatment of Advanced Gastric Cancer (INTEGRATE): A Multinational Placebo-Controlled Phase II Trial. J Clin Oncol 2016; 34:2728.
  96. Bang YJ, Kang YK, Kang WK, et al. Phase II study of sunitinib as second-line treatment for advanced gastric cancer. Invest New Drugs 2011; 29:1449.
  97. Sun W, Powell M, O'Dwyer PJ, et al. Phase II study of sorafenib in combination with docetaxel and cisplatin in the treatment of metastatic or advanced gastric and gastroesophageal junction adenocarcinoma: ECOG 5203. J Clin Oncol 2010; 28:2947.
  98. Yi JH, Lee J, Lee J, et al. Randomised phase II trial of docetaxel and sunitinib in patients with metastatic gastric cancer who were previously treated with fluoropyrimidine and platinum. Br J Cancer 2012; 106:1469.
  99. Cleary JM, Horick NK, McCleary NJ, et al. FOLFOX plus ziv-aflibercept or placebo in first-line metastatic esophagogastric adenocarcinoma: A double-blind, randomized, multicenter phase 2 trial. Cancer 2019; 125:2213.
Topic 121739 Version 27.0

References