Commentary: HER2-Positive EGA, Immunotherapy With Chemoradiation, and Lymph Node Metastasis in GC, December 2022

Article Type
Changed
Thu, 12/15/2022 - 16:28
Dr Uboha scans the journals so you don't have to!

 

A study by Hofheinz and colleagues evaluated whether targeting the human epidermal growth factor receptor 2 (HER2) pathway can improve outcomes in early-stage esophagogastric adenocarcinoma (EGA).1 About 15%-20% of EGA overexpress HER2. In the metastatic setting, anti-HER2 therapies have an established role. Trastuzumab in combination with chemotherapy has been part of standard treatment for these tumors for over a decade and now immunotherapy, based on the ongoing KEYNOTE-811 study, has been proven effective as well.2,3 However, prior attempts to effectively target HER2 in the early stage have not been successful.4

A phase 2 trial conducted by the AIO EGA Study Group evaluated the addition of trastuzumab and pertuzumab to FLOT (fluorouracil, leucovorin, oxaliplatin, docetaxel) chemotherapy in resectable HER2-positive EGA. The trial closed early, before planned full accrual, when the results of the JACOB trial, which evaluated the addition of pertuzumab in the metastatic setting, came out as negative.5 However, the results are still worth discussing here. A total of 81 patients were enrolled in this study (40 in the experimental arm and 41 in the chemotherapy-only control arm). Pathologic complete response was significantly improved with the addition of anti-HER2 therapy (35% vs 12%; P = .02). The rates of R0 resection and surgical complications were similar. Median disease-free survival was not reached in the experimental arm (26 months in the control arm). This study suggests that evaluation of anti-HER2 agents in combination with chemotherapy is warranted. Given the promising results of the KEYNOTE-811 study, future studies should consider incorporative immunotherapy as well.

The Neo-PLANET phase 2 study by Tang and colleagues evaluated the addition of camrelizumab (anti-PD1 antibody) to concurrent chemoradiation in the treatment of advanced EGA.6 The 36 patients enrolled in this study received induction chemotherapy with capecitabine and oxaliplatin followed by chemoradiation with the same chemotherapy backbone. Camrelizumab was added from the time of all chemotherapy initiation. The pathologic complete response observed (33.3%) compared favorably to historical references. No new safety signals were identified. Current standards for resectable EGA include adjuvant nivolumab in patients who have residual disease at the time of resection post chemoradiation. This study demonstrated that the addition of immunotherapy earlier in the treatment course is safe and possibly efficacious. The prospective randomized phase 2/3 ECOG-ACRIN 2174 study will evaluate the addition of nivolumab to chemoradiation, as well as the addition of ipilimumab to nivolumab in the adjuvant setting, and will answer the question regarding the benefits of immunotherapy used earlier in the course of disease in a prospective randomized manner (NCT03604991).

The study by de Jongh and colleagues evaluated the pattern of lymph node metastasis after neoadjuvant chemotherapy with relation to the location of the primary gastric tumor. Tumors from 212 patients who were previously enrolled in the Dutch LOGICA trial comparing laparoscopy vs open D2 gastrectomy were included in this analysis. Although the primary tumor location (proximal vs distal) was associated with a higher frequency of metastasis to certain lymph node groups, this relationship was not exclusive. As such, the extent of lymphadenectomy should not depend on the primary location of the tumor and is not affected by neoadjuvant chemotherapy.

Additional References

1. Hofheinz R-D, Merx K, Haag GM, et al. FLOT versus FLOT/trastuzumab/pertuzumab perioperative therapy of human epidermal growth factor receptor 2–positive resectable esophagogastric adenocarcinoma: A randomized phase II trial of the AIO EGA Study Group. J Clin Oncol. 2022;40:3750-3761. Doi: 10.1200/JCO.22.00380

2. Janjigian YY, Kawazoe A, Yañez P, et al. The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer. Nature. 2021;600:727-730. Doi: 10.1038/s41586-021-04161-3

3. Bang Y-J, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687-697. Doi: 10.1016/S0140-6736(10)61121-X

4. Safran HP, Winter K, Hilson D, et al. Trastuzumab with trimodality treatment for oesophageal adenocarcinoma with HER2 overexpression (NRG Oncology/RTOG 1010): A multicentre, randomised, phase 3 trial. Lancet Oncol. 2022;23:259-269. Doi: 10.1016/S1470-2045(21)00718-X

5. Tabernero J, Hoff PM, Shen L, et al. Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (JACOB): Final analysis of a double-blind, randomised, placebo-controlled phase 3 study. Lancet Oncol. 2018:19:1372-1384. Doi: 10.1016/S1470-2045(18)30481-9

6. Tang Z, Wang Y, Liu D, et al. The Neo-PLANET phase II trial of neoadjuvant camrelizumab plus concurrent chemoradiotherapy in locally advanced adenocarcinoma of stomach or gastroesophageal junction. Nat Commun 2022;13:6807. Doi: 10.1038/s41467-022-34403-5

 

 

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Publications
Topics
Sections
Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Dr Uboha scans the journals so you don't have to!
Dr Uboha scans the journals so you don't have to!

 

A study by Hofheinz and colleagues evaluated whether targeting the human epidermal growth factor receptor 2 (HER2) pathway can improve outcomes in early-stage esophagogastric adenocarcinoma (EGA).1 About 15%-20% of EGA overexpress HER2. In the metastatic setting, anti-HER2 therapies have an established role. Trastuzumab in combination with chemotherapy has been part of standard treatment for these tumors for over a decade and now immunotherapy, based on the ongoing KEYNOTE-811 study, has been proven effective as well.2,3 However, prior attempts to effectively target HER2 in the early stage have not been successful.4

A phase 2 trial conducted by the AIO EGA Study Group evaluated the addition of trastuzumab and pertuzumab to FLOT (fluorouracil, leucovorin, oxaliplatin, docetaxel) chemotherapy in resectable HER2-positive EGA. The trial closed early, before planned full accrual, when the results of the JACOB trial, which evaluated the addition of pertuzumab in the metastatic setting, came out as negative.5 However, the results are still worth discussing here. A total of 81 patients were enrolled in this study (40 in the experimental arm and 41 in the chemotherapy-only control arm). Pathologic complete response was significantly improved with the addition of anti-HER2 therapy (35% vs 12%; P = .02). The rates of R0 resection and surgical complications were similar. Median disease-free survival was not reached in the experimental arm (26 months in the control arm). This study suggests that evaluation of anti-HER2 agents in combination with chemotherapy is warranted. Given the promising results of the KEYNOTE-811 study, future studies should consider incorporative immunotherapy as well.

The Neo-PLANET phase 2 study by Tang and colleagues evaluated the addition of camrelizumab (anti-PD1 antibody) to concurrent chemoradiation in the treatment of advanced EGA.6 The 36 patients enrolled in this study received induction chemotherapy with capecitabine and oxaliplatin followed by chemoradiation with the same chemotherapy backbone. Camrelizumab was added from the time of all chemotherapy initiation. The pathologic complete response observed (33.3%) compared favorably to historical references. No new safety signals were identified. Current standards for resectable EGA include adjuvant nivolumab in patients who have residual disease at the time of resection post chemoradiation. This study demonstrated that the addition of immunotherapy earlier in the treatment course is safe and possibly efficacious. The prospective randomized phase 2/3 ECOG-ACRIN 2174 study will evaluate the addition of nivolumab to chemoradiation, as well as the addition of ipilimumab to nivolumab in the adjuvant setting, and will answer the question regarding the benefits of immunotherapy used earlier in the course of disease in a prospective randomized manner (NCT03604991).

The study by de Jongh and colleagues evaluated the pattern of lymph node metastasis after neoadjuvant chemotherapy with relation to the location of the primary gastric tumor. Tumors from 212 patients who were previously enrolled in the Dutch LOGICA trial comparing laparoscopy vs open D2 gastrectomy were included in this analysis. Although the primary tumor location (proximal vs distal) was associated with a higher frequency of metastasis to certain lymph node groups, this relationship was not exclusive. As such, the extent of lymphadenectomy should not depend on the primary location of the tumor and is not affected by neoadjuvant chemotherapy.

Additional References

1. Hofheinz R-D, Merx K, Haag GM, et al. FLOT versus FLOT/trastuzumab/pertuzumab perioperative therapy of human epidermal growth factor receptor 2–positive resectable esophagogastric adenocarcinoma: A randomized phase II trial of the AIO EGA Study Group. J Clin Oncol. 2022;40:3750-3761. Doi: 10.1200/JCO.22.00380

2. Janjigian YY, Kawazoe A, Yañez P, et al. The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer. Nature. 2021;600:727-730. Doi: 10.1038/s41586-021-04161-3

3. Bang Y-J, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687-697. Doi: 10.1016/S0140-6736(10)61121-X

4. Safran HP, Winter K, Hilson D, et al. Trastuzumab with trimodality treatment for oesophageal adenocarcinoma with HER2 overexpression (NRG Oncology/RTOG 1010): A multicentre, randomised, phase 3 trial. Lancet Oncol. 2022;23:259-269. Doi: 10.1016/S1470-2045(21)00718-X

5. Tabernero J, Hoff PM, Shen L, et al. Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (JACOB): Final analysis of a double-blind, randomised, placebo-controlled phase 3 study. Lancet Oncol. 2018:19:1372-1384. Doi: 10.1016/S1470-2045(18)30481-9

6. Tang Z, Wang Y, Liu D, et al. The Neo-PLANET phase II trial of neoadjuvant camrelizumab plus concurrent chemoradiotherapy in locally advanced adenocarcinoma of stomach or gastroesophageal junction. Nat Commun 2022;13:6807. Doi: 10.1038/s41467-022-34403-5

 

 

 

A study by Hofheinz and colleagues evaluated whether targeting the human epidermal growth factor receptor 2 (HER2) pathway can improve outcomes in early-stage esophagogastric adenocarcinoma (EGA).1 About 15%-20% of EGA overexpress HER2. In the metastatic setting, anti-HER2 therapies have an established role. Trastuzumab in combination with chemotherapy has been part of standard treatment for these tumors for over a decade and now immunotherapy, based on the ongoing KEYNOTE-811 study, has been proven effective as well.2,3 However, prior attempts to effectively target HER2 in the early stage have not been successful.4

A phase 2 trial conducted by the AIO EGA Study Group evaluated the addition of trastuzumab and pertuzumab to FLOT (fluorouracil, leucovorin, oxaliplatin, docetaxel) chemotherapy in resectable HER2-positive EGA. The trial closed early, before planned full accrual, when the results of the JACOB trial, which evaluated the addition of pertuzumab in the metastatic setting, came out as negative.5 However, the results are still worth discussing here. A total of 81 patients were enrolled in this study (40 in the experimental arm and 41 in the chemotherapy-only control arm). Pathologic complete response was significantly improved with the addition of anti-HER2 therapy (35% vs 12%; P = .02). The rates of R0 resection and surgical complications were similar. Median disease-free survival was not reached in the experimental arm (26 months in the control arm). This study suggests that evaluation of anti-HER2 agents in combination with chemotherapy is warranted. Given the promising results of the KEYNOTE-811 study, future studies should consider incorporative immunotherapy as well.

The Neo-PLANET phase 2 study by Tang and colleagues evaluated the addition of camrelizumab (anti-PD1 antibody) to concurrent chemoradiation in the treatment of advanced EGA.6 The 36 patients enrolled in this study received induction chemotherapy with capecitabine and oxaliplatin followed by chemoradiation with the same chemotherapy backbone. Camrelizumab was added from the time of all chemotherapy initiation. The pathologic complete response observed (33.3%) compared favorably to historical references. No new safety signals were identified. Current standards for resectable EGA include adjuvant nivolumab in patients who have residual disease at the time of resection post chemoradiation. This study demonstrated that the addition of immunotherapy earlier in the treatment course is safe and possibly efficacious. The prospective randomized phase 2/3 ECOG-ACRIN 2174 study will evaluate the addition of nivolumab to chemoradiation, as well as the addition of ipilimumab to nivolumab in the adjuvant setting, and will answer the question regarding the benefits of immunotherapy used earlier in the course of disease in a prospective randomized manner (NCT03604991).

The study by de Jongh and colleagues evaluated the pattern of lymph node metastasis after neoadjuvant chemotherapy with relation to the location of the primary gastric tumor. Tumors from 212 patients who were previously enrolled in the Dutch LOGICA trial comparing laparoscopy vs open D2 gastrectomy were included in this analysis. Although the primary tumor location (proximal vs distal) was associated with a higher frequency of metastasis to certain lymph node groups, this relationship was not exclusive. As such, the extent of lymphadenectomy should not depend on the primary location of the tumor and is not affected by neoadjuvant chemotherapy.

Additional References

1. Hofheinz R-D, Merx K, Haag GM, et al. FLOT versus FLOT/trastuzumab/pertuzumab perioperative therapy of human epidermal growth factor receptor 2–positive resectable esophagogastric adenocarcinoma: A randomized phase II trial of the AIO EGA Study Group. J Clin Oncol. 2022;40:3750-3761. Doi: 10.1200/JCO.22.00380

2. Janjigian YY, Kawazoe A, Yañez P, et al. The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer. Nature. 2021;600:727-730. Doi: 10.1038/s41586-021-04161-3

3. Bang Y-J, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687-697. Doi: 10.1016/S0140-6736(10)61121-X

4. Safran HP, Winter K, Hilson D, et al. Trastuzumab with trimodality treatment for oesophageal adenocarcinoma with HER2 overexpression (NRG Oncology/RTOG 1010): A multicentre, randomised, phase 3 trial. Lancet Oncol. 2022;23:259-269. Doi: 10.1016/S1470-2045(21)00718-X

5. Tabernero J, Hoff PM, Shen L, et al. Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (JACOB): Final analysis of a double-blind, randomised, placebo-controlled phase 3 study. Lancet Oncol. 2018:19:1372-1384. Doi: 10.1016/S1470-2045(18)30481-9

6. Tang Z, Wang Y, Liu D, et al. The Neo-PLANET phase II trial of neoadjuvant camrelizumab plus concurrent chemoradiotherapy in locally advanced adenocarcinoma of stomach or gastroesophageal junction. Nat Commun 2022;13:6807. Doi: 10.1038/s41467-022-34403-5

 

 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Gastric Cancer, December 2022
Gate On Date
Sun, 05/01/2022 - 20:00
Un-Gate On Date
Sun, 05/01/2022 - 20:00
Use ProPublica
CFC Schedule Remove Status
Sun, 05/01/2022 - 20:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
323615.9
Activity ID
84692
Product Name
Clinical Edge Journal Scan
Product ID
72
Supporter Name /ID
Erbitux [ 4139 ]

Commentary: Potential new treatments in gastroesophageal adenocarcinoma, November 2022

Article Type
Changed
Sat, 10/29/2022 - 16:51
Dr Uboha scans the journals so you don't have to!

 

 

Gastroesophageal adenocarcinomas are a diverse group of diseases that we are starting to reclassify more and more on the basis of biomarker profiles, such as microsatellite status, human epidermal growth factor receptor 2 (HER2) and programmed death-ligand 1 (PD-L1) positivity, and molecular signatures based on next-generation sequencing results. Recently, fibroblast growth factor receptor 2 (FGFR2), particularly the FGFR2b form, emerged as a potential future biomarker for treatment selection. FGFR2b as a biomarker is present in a significant proportion of upper gastrointestinal tumors.

The phase 2 FIGHT trial1 evaluated the role of bemarituzumab, an anti-FGFR2 antibody, in combination with chemotherapy during first-line treatment of advanced gastroesophageal adenocarcinoma. The primary endpoint of this trial was progression-free survival (PFS). This trial enrolled 155 patients with upper gastrointestinal tumors with FGFR2b overexpression (defined as at least 2+ by immunohistochemistry) or amplification on next-generation sequencing. About 30% of patients with HER2 nonpositive tumors (ie, those that would not qualify for treatment with the targeted agent trastuzumab) were eligible for participation. In the FIGHT trial, patients were randomized in a 1:1 ratio to receive either standard chemotherapy (folinic acid, fluorouracil, and oxaliplatin [FOLFOX]) or chemotherapy plus bemarituzumab. Patients in the experimental group were allowed to receive one dose of standard FOLFOX chemotherapy while biomarker testing was ongoing.

With a median follow-up time of 10.9 moths, PFS was numerically prolonged in the bemarituzumab group (9.5 vs 7.4 months), but it did not reach statistical significance (P = .073). Overall survival (OS) was improved in the experimental group (not reached vs 12.9 months; P = .027). With a longer follow-up of 12.5 months, in post hoc exploratory analysis, OS was significantly longer in the experimental group (19.2 vs 13.5 months; hazard ratio 0.60, P = .027). The rate of serious adverse events was similar between the two groups. However, it is important to note ocular toxicities associated with bemarituzumab treatment. Corneal adverse events were seen in 67% of patients in the experimental group, with 24% of patients experiencing grade 3 events. Moreover, 26% of patients discontinued bemarituzumab because of corneal adverse events.

Overall, this phase 2 trial demonstrated that FGFR2b is emerging as an important biomarker and target in patients with advanced gastroesophageal adenocarcinoma. Ongoing phase 3 trials (FORTITUDE-101 with FOLFOX [NCT05052801] and FORTITUDE-102 with FOLFOX and nivolumab [NCT05111626]) hopefully will confirm the early results seen in the FIGHT trial. Awareness and early attention to treatment-associated toxicities will be critical for the potential future incorporation of bemarituzumab into clinical practice.

A study by Ramos‐Santillan and colleagues explored whether the order of treatment modalities matter in the management of early-stage gastric cancer. Typically, perioperative chemotherapy (both neoadjuvant and adjuvant) is used during treatment of early-stage gastric cancer, which is usually defined as at least cT2N0 or cTxN+ disease. In this study, multivariable Cox regression analyses were performed on propensity score-matched cohorts. The study analyzed outcomes of 11,984 patients who were identified using the US National Cancer Database and treated between 2005 and 2014. The results revealed that patients who had stage I disease had better outcomes with upfront resection followed by adjuvant therapy. Patients with stage III disease did better with a neoadjuvant approach, whereas patients with stage II disease had similar outcomes regardless of chemotherapy timing. This research has the limitations inherent to the retrospective nature of the analysis and lack of prospective enrollment and controls. However, it does suggest that there may be a fraction of patients who should be treated with upfront resection. For incorporation of this change into standard practice, the question of therapy sequencing should be answered in a randomized prospective trial that incorporates the most updated systemic therapy (fluorouracil, leucovorin, oxaliplatin, and docetaxel [FLOT]) into its design.

Chemotherapy continues to play a critical role during first-line treatment of advanced esophageal and gastric adenocarcinoma. Triple chemotherapy regimens have been known to have increased efficacy in this setting, but their use has been limited by associated toxicities. A study by Nguyen and colleagues evaluated the TCX regimen (paclitaxel, carboplatin, and capecitabine) during first-line treatment of advanced gastric cancer. This regimen is similar to other triple chemotherapy regimens, such as FLOT and DCF (docetaxel, cisplatin, and fluorouracil), which have proven activity in this disease. This prospective phase 2 trial enrolled 83 patients. The median PFS (9.3 months) and OS (17 months) compared favorably with historical references. The regimen had expected adverse events, with cytopenias and fatigue being the most frequently reported. On the basis of the reported safety and efficacy, TCX has potential to be used as a chemotherapy backbone in future trials, but larger trials are needed to confirm the phase 2 trial results.

 

References

Wainberg ZA, Enzinger PC, Kang YK, et al. Bemarituzumab in patients with FGFR2b-selected gastric or gastro-oesophageal junction adenocarcinoma (FIGHT): A randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol. 2022 Oct 13. Doi: 10.1016/S1470-2045(22)00603-9

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Publications
Topics
Sections
Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Dr Uboha scans the journals so you don't have to!
Dr Uboha scans the journals so you don't have to!

 

 

Gastroesophageal adenocarcinomas are a diverse group of diseases that we are starting to reclassify more and more on the basis of biomarker profiles, such as microsatellite status, human epidermal growth factor receptor 2 (HER2) and programmed death-ligand 1 (PD-L1) positivity, and molecular signatures based on next-generation sequencing results. Recently, fibroblast growth factor receptor 2 (FGFR2), particularly the FGFR2b form, emerged as a potential future biomarker for treatment selection. FGFR2b as a biomarker is present in a significant proportion of upper gastrointestinal tumors.

The phase 2 FIGHT trial1 evaluated the role of bemarituzumab, an anti-FGFR2 antibody, in combination with chemotherapy during first-line treatment of advanced gastroesophageal adenocarcinoma. The primary endpoint of this trial was progression-free survival (PFS). This trial enrolled 155 patients with upper gastrointestinal tumors with FGFR2b overexpression (defined as at least 2+ by immunohistochemistry) or amplification on next-generation sequencing. About 30% of patients with HER2 nonpositive tumors (ie, those that would not qualify for treatment with the targeted agent trastuzumab) were eligible for participation. In the FIGHT trial, patients were randomized in a 1:1 ratio to receive either standard chemotherapy (folinic acid, fluorouracil, and oxaliplatin [FOLFOX]) or chemotherapy plus bemarituzumab. Patients in the experimental group were allowed to receive one dose of standard FOLFOX chemotherapy while biomarker testing was ongoing.

With a median follow-up time of 10.9 moths, PFS was numerically prolonged in the bemarituzumab group (9.5 vs 7.4 months), but it did not reach statistical significance (P = .073). Overall survival (OS) was improved in the experimental group (not reached vs 12.9 months; P = .027). With a longer follow-up of 12.5 months, in post hoc exploratory analysis, OS was significantly longer in the experimental group (19.2 vs 13.5 months; hazard ratio 0.60, P = .027). The rate of serious adverse events was similar between the two groups. However, it is important to note ocular toxicities associated with bemarituzumab treatment. Corneal adverse events were seen in 67% of patients in the experimental group, with 24% of patients experiencing grade 3 events. Moreover, 26% of patients discontinued bemarituzumab because of corneal adverse events.

Overall, this phase 2 trial demonstrated that FGFR2b is emerging as an important biomarker and target in patients with advanced gastroesophageal adenocarcinoma. Ongoing phase 3 trials (FORTITUDE-101 with FOLFOX [NCT05052801] and FORTITUDE-102 with FOLFOX and nivolumab [NCT05111626]) hopefully will confirm the early results seen in the FIGHT trial. Awareness and early attention to treatment-associated toxicities will be critical for the potential future incorporation of bemarituzumab into clinical practice.

A study by Ramos‐Santillan and colleagues explored whether the order of treatment modalities matter in the management of early-stage gastric cancer. Typically, perioperative chemotherapy (both neoadjuvant and adjuvant) is used during treatment of early-stage gastric cancer, which is usually defined as at least cT2N0 or cTxN+ disease. In this study, multivariable Cox regression analyses were performed on propensity score-matched cohorts. The study analyzed outcomes of 11,984 patients who were identified using the US National Cancer Database and treated between 2005 and 2014. The results revealed that patients who had stage I disease had better outcomes with upfront resection followed by adjuvant therapy. Patients with stage III disease did better with a neoadjuvant approach, whereas patients with stage II disease had similar outcomes regardless of chemotherapy timing. This research has the limitations inherent to the retrospective nature of the analysis and lack of prospective enrollment and controls. However, it does suggest that there may be a fraction of patients who should be treated with upfront resection. For incorporation of this change into standard practice, the question of therapy sequencing should be answered in a randomized prospective trial that incorporates the most updated systemic therapy (fluorouracil, leucovorin, oxaliplatin, and docetaxel [FLOT]) into its design.

Chemotherapy continues to play a critical role during first-line treatment of advanced esophageal and gastric adenocarcinoma. Triple chemotherapy regimens have been known to have increased efficacy in this setting, but their use has been limited by associated toxicities. A study by Nguyen and colleagues evaluated the TCX regimen (paclitaxel, carboplatin, and capecitabine) during first-line treatment of advanced gastric cancer. This regimen is similar to other triple chemotherapy regimens, such as FLOT and DCF (docetaxel, cisplatin, and fluorouracil), which have proven activity in this disease. This prospective phase 2 trial enrolled 83 patients. The median PFS (9.3 months) and OS (17 months) compared favorably with historical references. The regimen had expected adverse events, with cytopenias and fatigue being the most frequently reported. On the basis of the reported safety and efficacy, TCX has potential to be used as a chemotherapy backbone in future trials, but larger trials are needed to confirm the phase 2 trial results.

 

References

Wainberg ZA, Enzinger PC, Kang YK, et al. Bemarituzumab in patients with FGFR2b-selected gastric or gastro-oesophageal junction adenocarcinoma (FIGHT): A randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol. 2022 Oct 13. Doi: 10.1016/S1470-2045(22)00603-9

 

 

Gastroesophageal adenocarcinomas are a diverse group of diseases that we are starting to reclassify more and more on the basis of biomarker profiles, such as microsatellite status, human epidermal growth factor receptor 2 (HER2) and programmed death-ligand 1 (PD-L1) positivity, and molecular signatures based on next-generation sequencing results. Recently, fibroblast growth factor receptor 2 (FGFR2), particularly the FGFR2b form, emerged as a potential future biomarker for treatment selection. FGFR2b as a biomarker is present in a significant proportion of upper gastrointestinal tumors.

The phase 2 FIGHT trial1 evaluated the role of bemarituzumab, an anti-FGFR2 antibody, in combination with chemotherapy during first-line treatment of advanced gastroesophageal adenocarcinoma. The primary endpoint of this trial was progression-free survival (PFS). This trial enrolled 155 patients with upper gastrointestinal tumors with FGFR2b overexpression (defined as at least 2+ by immunohistochemistry) or amplification on next-generation sequencing. About 30% of patients with HER2 nonpositive tumors (ie, those that would not qualify for treatment with the targeted agent trastuzumab) were eligible for participation. In the FIGHT trial, patients were randomized in a 1:1 ratio to receive either standard chemotherapy (folinic acid, fluorouracil, and oxaliplatin [FOLFOX]) or chemotherapy plus bemarituzumab. Patients in the experimental group were allowed to receive one dose of standard FOLFOX chemotherapy while biomarker testing was ongoing.

With a median follow-up time of 10.9 moths, PFS was numerically prolonged in the bemarituzumab group (9.5 vs 7.4 months), but it did not reach statistical significance (P = .073). Overall survival (OS) was improved in the experimental group (not reached vs 12.9 months; P = .027). With a longer follow-up of 12.5 months, in post hoc exploratory analysis, OS was significantly longer in the experimental group (19.2 vs 13.5 months; hazard ratio 0.60, P = .027). The rate of serious adverse events was similar between the two groups. However, it is important to note ocular toxicities associated with bemarituzumab treatment. Corneal adverse events were seen in 67% of patients in the experimental group, with 24% of patients experiencing grade 3 events. Moreover, 26% of patients discontinued bemarituzumab because of corneal adverse events.

Overall, this phase 2 trial demonstrated that FGFR2b is emerging as an important biomarker and target in patients with advanced gastroesophageal adenocarcinoma. Ongoing phase 3 trials (FORTITUDE-101 with FOLFOX [NCT05052801] and FORTITUDE-102 with FOLFOX and nivolumab [NCT05111626]) hopefully will confirm the early results seen in the FIGHT trial. Awareness and early attention to treatment-associated toxicities will be critical for the potential future incorporation of bemarituzumab into clinical practice.

A study by Ramos‐Santillan and colleagues explored whether the order of treatment modalities matter in the management of early-stage gastric cancer. Typically, perioperative chemotherapy (both neoadjuvant and adjuvant) is used during treatment of early-stage gastric cancer, which is usually defined as at least cT2N0 or cTxN+ disease. In this study, multivariable Cox regression analyses were performed on propensity score-matched cohorts. The study analyzed outcomes of 11,984 patients who were identified using the US National Cancer Database and treated between 2005 and 2014. The results revealed that patients who had stage I disease had better outcomes with upfront resection followed by adjuvant therapy. Patients with stage III disease did better with a neoadjuvant approach, whereas patients with stage II disease had similar outcomes regardless of chemotherapy timing. This research has the limitations inherent to the retrospective nature of the analysis and lack of prospective enrollment and controls. However, it does suggest that there may be a fraction of patients who should be treated with upfront resection. For incorporation of this change into standard practice, the question of therapy sequencing should be answered in a randomized prospective trial that incorporates the most updated systemic therapy (fluorouracil, leucovorin, oxaliplatin, and docetaxel [FLOT]) into its design.

Chemotherapy continues to play a critical role during first-line treatment of advanced esophageal and gastric adenocarcinoma. Triple chemotherapy regimens have been known to have increased efficacy in this setting, but their use has been limited by associated toxicities. A study by Nguyen and colleagues evaluated the TCX regimen (paclitaxel, carboplatin, and capecitabine) during first-line treatment of advanced gastric cancer. This regimen is similar to other triple chemotherapy regimens, such as FLOT and DCF (docetaxel, cisplatin, and fluorouracil), which have proven activity in this disease. This prospective phase 2 trial enrolled 83 patients. The median PFS (9.3 months) and OS (17 months) compared favorably with historical references. The regimen had expected adverse events, with cytopenias and fatigue being the most frequently reported. On the basis of the reported safety and efficacy, TCX has potential to be used as a chemotherapy backbone in future trials, but larger trials are needed to confirm the phase 2 trial results.

 

References

Wainberg ZA, Enzinger PC, Kang YK, et al. Bemarituzumab in patients with FGFR2b-selected gastric or gastro-oesophageal junction adenocarcinoma (FIGHT): A randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol. 2022 Oct 13. Doi: 10.1016/S1470-2045(22)00603-9

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Gastric Cancer, November 2022
Gate On Date
Sun, 05/01/2022 - 20:00
Un-Gate On Date
Sun, 05/01/2022 - 20:00
Use ProPublica
CFC Schedule Remove Status
Sun, 05/01/2022 - 20:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
323615.9
Activity ID
84692
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Erbitux [ 4139 ]

Commentary: Biomarkers and Chemotherapy in Gastric Cancer, October 2022

Article Type
Changed
Mon, 10/03/2022 - 14:10
Dr Uboha scans the journals so you don't have to!

 

Immune checkpoint inhibitors in combination with chemotherapy are part of standard treatment for patients with advanced gastroesophageal adenocarcinoma. However, not all patients benefit from immunotherapy addition. Programmed death-ligand 1 (PD-L1) expression has emerged as a useful, yet imperfect, biomarker for identifying patients who are most likely to benefit from the addition of immunotherapy. There is a need to better identify patients with higher chances of responding to these therapies, as well as a need to augment the activity that we are seeing with current approaches. Tumor mutational burden (TMB) has been previously identified as a predictive biomarker of response to immunotherapy. It is not widely used in clinical practice when treating patients with upper gastrointestinal cancers though, and it has not been extensively evaluated in this disease.

The study by Lee and colleagues evaluated the association between TMB and pembrolizumab response in patients treated in the phase 3 KEYNOTE-62 study. This was a prespecified exploratory analysis, which included 306 of 763 patients, based on TMB data availability. Although there was association between high TMB (cut-off defined as 10 mut/Mb), the benefit from pembrolizumab was most pronounced in patients with microsatellite-unstable tumors. When the analysis was limited to microsatellite-stable tumors with high TMB, the signal was attenuated and was only detected when immunotherapy was combined with chemotherapy.

Ultimately, although TMB is a predictive biomarker of response to immunotherapy, it is unlikely to significantly alter our approach to treatment of upper gastrointestinal cancers, especially when PD-L1 testing and microsatellite testing are more readily available and do not require more time- and resource-consuming next-generation sequencing when selecting first-line treatments.

Perioperative chemotherapy plays a critical role in the management of patients with early-stage gastric cancer. In the United States, perioperative FLOT (5-fluorouracil, oxaliplatin, and docetaxel) chemotherapy is the standard approach to this disease, with four cycles administered before and four cycles administered after resection.

In Korea, adjuvant chemotherapy is typically used, with 1 year of S-1 chemotherapy or 6 months of CAPOX (capecitabine/oxaliplatin) chemotherapy used most frequently. Kim and colleagues analyzed whether a shorter duration of chemotherapy (either S-1 or CAPOX) had similar outcomes. In a retrospective analysis of 20,552 patients, which included 13,614 patients who received S-1 and 6938 patients who received CAPOX, overall survival was worse in those patients who received a shorter duration of adjuvant treatment. Although this study is not directly applicable to how we approach treatment of early-stage gastric cancer in the United States, it does suggest that any modification in the duration of perioperative chemotherapy should be evaluated in prospective clinical trials, similarly to recent investigations regarding the duration on adjuvant chemotherapy in colon cancer.

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Publications
Topics
Sections
Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Dr Uboha scans the journals so you don't have to!
Dr Uboha scans the journals so you don't have to!

 

Immune checkpoint inhibitors in combination with chemotherapy are part of standard treatment for patients with advanced gastroesophageal adenocarcinoma. However, not all patients benefit from immunotherapy addition. Programmed death-ligand 1 (PD-L1) expression has emerged as a useful, yet imperfect, biomarker for identifying patients who are most likely to benefit from the addition of immunotherapy. There is a need to better identify patients with higher chances of responding to these therapies, as well as a need to augment the activity that we are seeing with current approaches. Tumor mutational burden (TMB) has been previously identified as a predictive biomarker of response to immunotherapy. It is not widely used in clinical practice when treating patients with upper gastrointestinal cancers though, and it has not been extensively evaluated in this disease.

The study by Lee and colleagues evaluated the association between TMB and pembrolizumab response in patients treated in the phase 3 KEYNOTE-62 study. This was a prespecified exploratory analysis, which included 306 of 763 patients, based on TMB data availability. Although there was association between high TMB (cut-off defined as 10 mut/Mb), the benefit from pembrolizumab was most pronounced in patients with microsatellite-unstable tumors. When the analysis was limited to microsatellite-stable tumors with high TMB, the signal was attenuated and was only detected when immunotherapy was combined with chemotherapy.

Ultimately, although TMB is a predictive biomarker of response to immunotherapy, it is unlikely to significantly alter our approach to treatment of upper gastrointestinal cancers, especially when PD-L1 testing and microsatellite testing are more readily available and do not require more time- and resource-consuming next-generation sequencing when selecting first-line treatments.

Perioperative chemotherapy plays a critical role in the management of patients with early-stage gastric cancer. In the United States, perioperative FLOT (5-fluorouracil, oxaliplatin, and docetaxel) chemotherapy is the standard approach to this disease, with four cycles administered before and four cycles administered after resection.

In Korea, adjuvant chemotherapy is typically used, with 1 year of S-1 chemotherapy or 6 months of CAPOX (capecitabine/oxaliplatin) chemotherapy used most frequently. Kim and colleagues analyzed whether a shorter duration of chemotherapy (either S-1 or CAPOX) had similar outcomes. In a retrospective analysis of 20,552 patients, which included 13,614 patients who received S-1 and 6938 patients who received CAPOX, overall survival was worse in those patients who received a shorter duration of adjuvant treatment. Although this study is not directly applicable to how we approach treatment of early-stage gastric cancer in the United States, it does suggest that any modification in the duration of perioperative chemotherapy should be evaluated in prospective clinical trials, similarly to recent investigations regarding the duration on adjuvant chemotherapy in colon cancer.

 

Immune checkpoint inhibitors in combination with chemotherapy are part of standard treatment for patients with advanced gastroesophageal adenocarcinoma. However, not all patients benefit from immunotherapy addition. Programmed death-ligand 1 (PD-L1) expression has emerged as a useful, yet imperfect, biomarker for identifying patients who are most likely to benefit from the addition of immunotherapy. There is a need to better identify patients with higher chances of responding to these therapies, as well as a need to augment the activity that we are seeing with current approaches. Tumor mutational burden (TMB) has been previously identified as a predictive biomarker of response to immunotherapy. It is not widely used in clinical practice when treating patients with upper gastrointestinal cancers though, and it has not been extensively evaluated in this disease.

The study by Lee and colleagues evaluated the association between TMB and pembrolizumab response in patients treated in the phase 3 KEYNOTE-62 study. This was a prespecified exploratory analysis, which included 306 of 763 patients, based on TMB data availability. Although there was association between high TMB (cut-off defined as 10 mut/Mb), the benefit from pembrolizumab was most pronounced in patients with microsatellite-unstable tumors. When the analysis was limited to microsatellite-stable tumors with high TMB, the signal was attenuated and was only detected when immunotherapy was combined with chemotherapy.

Ultimately, although TMB is a predictive biomarker of response to immunotherapy, it is unlikely to significantly alter our approach to treatment of upper gastrointestinal cancers, especially when PD-L1 testing and microsatellite testing are more readily available and do not require more time- and resource-consuming next-generation sequencing when selecting first-line treatments.

Perioperative chemotherapy plays a critical role in the management of patients with early-stage gastric cancer. In the United States, perioperative FLOT (5-fluorouracil, oxaliplatin, and docetaxel) chemotherapy is the standard approach to this disease, with four cycles administered before and four cycles administered after resection.

In Korea, adjuvant chemotherapy is typically used, with 1 year of S-1 chemotherapy or 6 months of CAPOX (capecitabine/oxaliplatin) chemotherapy used most frequently. Kim and colleagues analyzed whether a shorter duration of chemotherapy (either S-1 or CAPOX) had similar outcomes. In a retrospective analysis of 20,552 patients, which included 13,614 patients who received S-1 and 6938 patients who received CAPOX, overall survival was worse in those patients who received a shorter duration of adjuvant treatment. Although this study is not directly applicable to how we approach treatment of early-stage gastric cancer in the United States, it does suggest that any modification in the duration of perioperative chemotherapy should be evaluated in prospective clinical trials, similarly to recent investigations regarding the duration on adjuvant chemotherapy in colon cancer.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Gastric Caner, October 2022
Gate On Date
Sun, 05/01/2022 - 20:00
Un-Gate On Date
Sun, 05/01/2022 - 20:00
Use ProPublica
CFC Schedule Remove Status
Sun, 05/01/2022 - 20:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
323615.9
Activity ID
84692
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Erbitux [ 4139 ]

Commentary: Gastric Cancer Surgery Trials, September 2022

Article Type
Changed
Thu, 09/01/2022 - 11:30
Dr Uboha scans the journals so you don't have to!

 

Surgical resection plays a critical role in the management of early-stage gastric cancer. Depending on the tumor stage and location, there are different surgical approaches. Complications associated with surgical resection can significantly affect quality of life and ability to receive subsequent treatment. With recent advances in minimally invasive approaches, laparoscopic resections are emerging as an attractive option for patients undergoing oncologic surgeries.

The KLASS-02 trial was a multicenter, randomized, controlled, noninferiority clinical trial, which enrolled 1050 patients with locally advanced gastric cancer. Of the enrolled patients, 974 patients underwent R0 resection either by laparoscopic (n = 492) or open (n = 482) distal gastrectomy. In the previous readout of this study with 3 years of follow-up, laparoscopic distal gastrectomy had noninferior oncologic outcomes compared with open surgery for locally advanced gastric cancer. Son and colleagues are now reporting 5-year follow-up results. Overall survival (OS; 88.9% vs 88.7%; P = .30) and relapse-free survival (79.5% vs 81.1%; P = .658) rates were similar in both surgical groups. The pattern of recurrences was similar between the two groups as well, with peroneal (42.1% of patients) and hematogenous (20.8%) being the most frequent ones. However, patients who underwent laparoscopic vs open distal gastrectomy had a significantly lower late complication rate (6.5% vs 11.0%; P = .01). This study demonstrates that laparoscopic distal gastrectomy is an appropriate alternative to open distal gastrectomy and should be offered to patients who are treated in centers with experience in performing these types of operations.

Peritoneal metastasis is a common site for the spread of gastric cancer. As such, the role of hyperthermic intraperitoneal chemotherapy (HIPEC) in the management of this disease has been explored in a number of studies. The effectiveness of prophylactic HIPEC during resection of early-stage gastric cancer remains unknown.

Shen and colleagues conducted a propensity score-matching analysis looking at the efficacy and safety of HIPEC in this setting. The study evaluated outcomes of 395 patients with locally advanced gastric cancer who underwent resection with (n = 146) or without HIPEC (n = 248). In the HIPEC group, OS compared favorably to the surgery-only group (69.9% vs 40.8%, P = .049) and 2-year relapse-free survival was higher with HIPEC (60.7% vs 31.6%, P = .049).

Previously, the CYTO-CHIP propensity score analysis study performed in France demonstrated that HIPEC in addition to cytoreductive surgery resulted in improved OS in patients with advanced gastric cancer and peritoneal metastasis compared to cytoreduction surgery alone.1 However, with both of these reports, interpretation of the results carries inherent limitations that are associated with retrospective study design. Prior prospective studies, on the other hand, had mixed results. An ongoing phase 3 prospective study of D2 resection and HIPEC in locally advanced gastric carcinoma (GASTRICHIP) will hopefully provide a definitive answer regarding the benefit of HIPEC in early-stage gastric cancer management (NCT01882933). Ultimately, going forward, the role of HIPEC in early-stage disease will need to be examined in a prospective study with carefully selected patients, using the latest biomarkers and systemic therapies.

Mismatch repair protein deficient (dMMR) or microsatellite unstable gastric cancer (MSI-H) have distinct biologic behaviors and treatment responses. They are much more responsive to immune checkpoint inhibitors in the metastatic setting. In early-stage disease, exploratory analysis of patients with MSI-H tumors previously enrolled in the perioperative MAGIC trial, revealed that patients with MSI-H tumors had a better prognosis when treated with surgery alone, and they potentially experienced detrimental effects from chemotherapy.2

The GERCOR NEONIPIGA single-arm phase 2 study enrolled 32 patients with resectable dMMR/MSI-H gastric and gastroesophageal junction tumors.3 Patients were treated with 240-mg neoadjuvant nivolumab once every 2 weeks six times and 1-mg/kg ipilimumab once every 6 weeks twice, followed by surgery and 480-mg adjuvant nivolumab once every 4 weeks nine times. Twenty-nine patients underwent resection. All resections were with negative margins (R0). Pathologic complete response was seen in 17 (58.6%) of patients. As of the February 2022 data cutoff, with a median duration of follow-up of 14.9 months, 30 out of 31 patients with early-stage disease remained alive and without recurrence or progression (one evaluable patient had metastatic disease).

These results certainly support further investigation of immunotherapy use in this patient population. However, in the absence of prospective randomized data, the combination of fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT) remains the standard of care in those who can tolerate it. For patients with MSI-H tumors who are not candidates for combination chemotherapy or whose tumors are progressing on chemotherapy, neoadjuvant immunotherapy is certainly a good option to consider.

 

Additional References

1. Bonnot P-E et al. Cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy for gastric cancer with peritoneal metastases (CYTO-CHIP study): A propensity score analysis. J Clin Oncol. 2019;37:2028-2040. Doi: 10.1200/JCO.18.01688

2. Smyth EC et al. Mismatch repair deficiency, microsatellite instability, and survival: An exploratory analysis of the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) trial. JAMA Oncol. 2017;3:1197-1203. Doi: 10.1001/jamaoncol.2016.6762

3. André T et al. Neoadjuvant nivolumab plus ipilimumab and adjuvant nivolumab in localized deficient mismatch repair/microsatellite instability-high gastric or esophagogastric junction adenocarcinoma: The GERCOR NEONIPIGA phase II study. J Clin Oncol. 2022 (Aug 15. Doi: 10.1200/JCO.22.00686

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Publications
Topics
Sections
Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Dr Uboha scans the journals so you don't have to!
Dr Uboha scans the journals so you don't have to!

 

Surgical resection plays a critical role in the management of early-stage gastric cancer. Depending on the tumor stage and location, there are different surgical approaches. Complications associated with surgical resection can significantly affect quality of life and ability to receive subsequent treatment. With recent advances in minimally invasive approaches, laparoscopic resections are emerging as an attractive option for patients undergoing oncologic surgeries.

The KLASS-02 trial was a multicenter, randomized, controlled, noninferiority clinical trial, which enrolled 1050 patients with locally advanced gastric cancer. Of the enrolled patients, 974 patients underwent R0 resection either by laparoscopic (n = 492) or open (n = 482) distal gastrectomy. In the previous readout of this study with 3 years of follow-up, laparoscopic distal gastrectomy had noninferior oncologic outcomes compared with open surgery for locally advanced gastric cancer. Son and colleagues are now reporting 5-year follow-up results. Overall survival (OS; 88.9% vs 88.7%; P = .30) and relapse-free survival (79.5% vs 81.1%; P = .658) rates were similar in both surgical groups. The pattern of recurrences was similar between the two groups as well, with peroneal (42.1% of patients) and hematogenous (20.8%) being the most frequent ones. However, patients who underwent laparoscopic vs open distal gastrectomy had a significantly lower late complication rate (6.5% vs 11.0%; P = .01). This study demonstrates that laparoscopic distal gastrectomy is an appropriate alternative to open distal gastrectomy and should be offered to patients who are treated in centers with experience in performing these types of operations.

Peritoneal metastasis is a common site for the spread of gastric cancer. As such, the role of hyperthermic intraperitoneal chemotherapy (HIPEC) in the management of this disease has been explored in a number of studies. The effectiveness of prophylactic HIPEC during resection of early-stage gastric cancer remains unknown.

Shen and colleagues conducted a propensity score-matching analysis looking at the efficacy and safety of HIPEC in this setting. The study evaluated outcomes of 395 patients with locally advanced gastric cancer who underwent resection with (n = 146) or without HIPEC (n = 248). In the HIPEC group, OS compared favorably to the surgery-only group (69.9% vs 40.8%, P = .049) and 2-year relapse-free survival was higher with HIPEC (60.7% vs 31.6%, P = .049).

Previously, the CYTO-CHIP propensity score analysis study performed in France demonstrated that HIPEC in addition to cytoreductive surgery resulted in improved OS in patients with advanced gastric cancer and peritoneal metastasis compared to cytoreduction surgery alone.1 However, with both of these reports, interpretation of the results carries inherent limitations that are associated with retrospective study design. Prior prospective studies, on the other hand, had mixed results. An ongoing phase 3 prospective study of D2 resection and HIPEC in locally advanced gastric carcinoma (GASTRICHIP) will hopefully provide a definitive answer regarding the benefit of HIPEC in early-stage gastric cancer management (NCT01882933). Ultimately, going forward, the role of HIPEC in early-stage disease will need to be examined in a prospective study with carefully selected patients, using the latest biomarkers and systemic therapies.

Mismatch repair protein deficient (dMMR) or microsatellite unstable gastric cancer (MSI-H) have distinct biologic behaviors and treatment responses. They are much more responsive to immune checkpoint inhibitors in the metastatic setting. In early-stage disease, exploratory analysis of patients with MSI-H tumors previously enrolled in the perioperative MAGIC trial, revealed that patients with MSI-H tumors had a better prognosis when treated with surgery alone, and they potentially experienced detrimental effects from chemotherapy.2

The GERCOR NEONIPIGA single-arm phase 2 study enrolled 32 patients with resectable dMMR/MSI-H gastric and gastroesophageal junction tumors.3 Patients were treated with 240-mg neoadjuvant nivolumab once every 2 weeks six times and 1-mg/kg ipilimumab once every 6 weeks twice, followed by surgery and 480-mg adjuvant nivolumab once every 4 weeks nine times. Twenty-nine patients underwent resection. All resections were with negative margins (R0). Pathologic complete response was seen in 17 (58.6%) of patients. As of the February 2022 data cutoff, with a median duration of follow-up of 14.9 months, 30 out of 31 patients with early-stage disease remained alive and without recurrence or progression (one evaluable patient had metastatic disease).

These results certainly support further investigation of immunotherapy use in this patient population. However, in the absence of prospective randomized data, the combination of fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT) remains the standard of care in those who can tolerate it. For patients with MSI-H tumors who are not candidates for combination chemotherapy or whose tumors are progressing on chemotherapy, neoadjuvant immunotherapy is certainly a good option to consider.

 

Additional References

1. Bonnot P-E et al. Cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy for gastric cancer with peritoneal metastases (CYTO-CHIP study): A propensity score analysis. J Clin Oncol. 2019;37:2028-2040. Doi: 10.1200/JCO.18.01688

2. Smyth EC et al. Mismatch repair deficiency, microsatellite instability, and survival: An exploratory analysis of the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) trial. JAMA Oncol. 2017;3:1197-1203. Doi: 10.1001/jamaoncol.2016.6762

3. André T et al. Neoadjuvant nivolumab plus ipilimumab and adjuvant nivolumab in localized deficient mismatch repair/microsatellite instability-high gastric or esophagogastric junction adenocarcinoma: The GERCOR NEONIPIGA phase II study. J Clin Oncol. 2022 (Aug 15. Doi: 10.1200/JCO.22.00686

 

Surgical resection plays a critical role in the management of early-stage gastric cancer. Depending on the tumor stage and location, there are different surgical approaches. Complications associated with surgical resection can significantly affect quality of life and ability to receive subsequent treatment. With recent advances in minimally invasive approaches, laparoscopic resections are emerging as an attractive option for patients undergoing oncologic surgeries.

The KLASS-02 trial was a multicenter, randomized, controlled, noninferiority clinical trial, which enrolled 1050 patients with locally advanced gastric cancer. Of the enrolled patients, 974 patients underwent R0 resection either by laparoscopic (n = 492) or open (n = 482) distal gastrectomy. In the previous readout of this study with 3 years of follow-up, laparoscopic distal gastrectomy had noninferior oncologic outcomes compared with open surgery for locally advanced gastric cancer. Son and colleagues are now reporting 5-year follow-up results. Overall survival (OS; 88.9% vs 88.7%; P = .30) and relapse-free survival (79.5% vs 81.1%; P = .658) rates were similar in both surgical groups. The pattern of recurrences was similar between the two groups as well, with peroneal (42.1% of patients) and hematogenous (20.8%) being the most frequent ones. However, patients who underwent laparoscopic vs open distal gastrectomy had a significantly lower late complication rate (6.5% vs 11.0%; P = .01). This study demonstrates that laparoscopic distal gastrectomy is an appropriate alternative to open distal gastrectomy and should be offered to patients who are treated in centers with experience in performing these types of operations.

Peritoneal metastasis is a common site for the spread of gastric cancer. As such, the role of hyperthermic intraperitoneal chemotherapy (HIPEC) in the management of this disease has been explored in a number of studies. The effectiveness of prophylactic HIPEC during resection of early-stage gastric cancer remains unknown.

Shen and colleagues conducted a propensity score-matching analysis looking at the efficacy and safety of HIPEC in this setting. The study evaluated outcomes of 395 patients with locally advanced gastric cancer who underwent resection with (n = 146) or without HIPEC (n = 248). In the HIPEC group, OS compared favorably to the surgery-only group (69.9% vs 40.8%, P = .049) and 2-year relapse-free survival was higher with HIPEC (60.7% vs 31.6%, P = .049).

Previously, the CYTO-CHIP propensity score analysis study performed in France demonstrated that HIPEC in addition to cytoreductive surgery resulted in improved OS in patients with advanced gastric cancer and peritoneal metastasis compared to cytoreduction surgery alone.1 However, with both of these reports, interpretation of the results carries inherent limitations that are associated with retrospective study design. Prior prospective studies, on the other hand, had mixed results. An ongoing phase 3 prospective study of D2 resection and HIPEC in locally advanced gastric carcinoma (GASTRICHIP) will hopefully provide a definitive answer regarding the benefit of HIPEC in early-stage gastric cancer management (NCT01882933). Ultimately, going forward, the role of HIPEC in early-stage disease will need to be examined in a prospective study with carefully selected patients, using the latest biomarkers and systemic therapies.

Mismatch repair protein deficient (dMMR) or microsatellite unstable gastric cancer (MSI-H) have distinct biologic behaviors and treatment responses. They are much more responsive to immune checkpoint inhibitors in the metastatic setting. In early-stage disease, exploratory analysis of patients with MSI-H tumors previously enrolled in the perioperative MAGIC trial, revealed that patients with MSI-H tumors had a better prognosis when treated with surgery alone, and they potentially experienced detrimental effects from chemotherapy.2

The GERCOR NEONIPIGA single-arm phase 2 study enrolled 32 patients with resectable dMMR/MSI-H gastric and gastroesophageal junction tumors.3 Patients were treated with 240-mg neoadjuvant nivolumab once every 2 weeks six times and 1-mg/kg ipilimumab once every 6 weeks twice, followed by surgery and 480-mg adjuvant nivolumab once every 4 weeks nine times. Twenty-nine patients underwent resection. All resections were with negative margins (R0). Pathologic complete response was seen in 17 (58.6%) of patients. As of the February 2022 data cutoff, with a median duration of follow-up of 14.9 months, 30 out of 31 patients with early-stage disease remained alive and without recurrence or progression (one evaluable patient had metastatic disease).

These results certainly support further investigation of immunotherapy use in this patient population. However, in the absence of prospective randomized data, the combination of fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT) remains the standard of care in those who can tolerate it. For patients with MSI-H tumors who are not candidates for combination chemotherapy or whose tumors are progressing on chemotherapy, neoadjuvant immunotherapy is certainly a good option to consider.

 

Additional References

1. Bonnot P-E et al. Cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy for gastric cancer with peritoneal metastases (CYTO-CHIP study): A propensity score analysis. J Clin Oncol. 2019;37:2028-2040. Doi: 10.1200/JCO.18.01688

2. Smyth EC et al. Mismatch repair deficiency, microsatellite instability, and survival: An exploratory analysis of the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) trial. JAMA Oncol. 2017;3:1197-1203. Doi: 10.1001/jamaoncol.2016.6762

3. André T et al. Neoadjuvant nivolumab plus ipilimumab and adjuvant nivolumab in localized deficient mismatch repair/microsatellite instability-high gastric or esophagogastric junction adenocarcinoma: The GERCOR NEONIPIGA phase II study. J Clin Oncol. 2022 (Aug 15. Doi: 10.1200/JCO.22.00686

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan Commentary: Gastric Cancer, September 2022
Gate On Date
Sun, 05/01/2022 - 20:00
Un-Gate On Date
Sun, 05/01/2022 - 20:00
Use ProPublica
CFC Schedule Remove Status
Sun, 05/01/2022 - 20:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
323615.9
Activity ID
84692
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Erbitux [ 4139 ]

Commentary: Treating Gastric Cancer Subtypes, August 2022

Article Type
Changed
Fri, 08/12/2022 - 11:00
Dr Uboha scans the journals so you don't have to!

 

Patients with stage II or III gastric cancer are treated with surgical resection and perioperative chemotherapy. Platinum agents have established activity in this disease. Combination chemotherapy FLOT (5-fluorouracil, oxaliplatin, and docetaxel) is now standard perioperative treatment for resectable gastric cancer.1 The study by Slagter and colleagues evaluated whether cisplatin was noninferior to oxaliplatin when used in the treatment of early-stage gastric cancer. Prior to the incorporation of FLOT into standard treatment practice, patients were treated with ECX (epirubicin, cisplatin, and docetaxel), as per the MAGIC trial.2 In the metastatic setting, chemotherapy regimens with either cisplatin or oxaliplatin as a choice of platinum agent have comparable activity against these tumors. Oxaliplatin activity has been shown to be noninferior to cisplatin in the randomized REAL2 trial in metastatic setting.3

The study by Slagter and colleagues is a post hoc analysis of 781 patients with resectable gastric cancer who were enrolled in the CRITICS trial. This analysis demonstrated that chemotherapy regimens containing oxaliplatin and cisplatin had comparable 5-year overall survival rates. Not surprisingly, oxaliplatin was associated with higher neurotoxicity. Based on this analysis, it is likely safe to conclude that, just as in the advanced setting, cisplatin and oxaliplatin have similar activity in early-stage disease.

Mismatch repair protein deficient or microsatellite unstable gastric cancer (MSI-H) represent unique subtypes of gastric cancer, with distinct biologic behaviors and treatment responses. The efficacy of chemotherapy in patients with early-stage MSI-H tumors has been questioned previously. Similar to MSI-H colorectal cancers, the benefit of chemotherapy in resectable MSI-H gastric and esophagogastric junction tumors appears to be less robust than in microsatellite stable (MSS) tumors. In the exploratory analysis of patients with MSI-H tumors enrolled in the perioperative MAGIC trial, patients with MSI-H tumors had better prognosis when treated with surgery alone and potentially experienced detrimental effects from chemotherapy.4 The retrospective analysis by Vos and colleagues adds to the body of knowledge about early-stage MSI-H gastric cancers. They evaluated 535 patients with early-stage disease who were treated with surgery alone or surgery plus perioperative therapy between 2000 and 2018. The overall survival in 82 patients with MSI-H tumors was 20% better than in those with MSS disease. This favorable outcome was seen irrespective of whether chemotherapy was given. Though these results suggest that chemotherapy may not be necessary in the treatment of these tumors and there are emerging data regarding the activity of immune checkpoint inhibitors in this setting, these results should definitely be investigated further in prospective studies.5 However, in the absence of prospective randomized data, it is difficult to recommend deviating from the established standard of care with FLOT, especially for patients undergoing curative intent treatment.

A study by Yukami and colleagues evaluated whether the presence of liver metastasis, which have been shown to be enriched in immunosuppressive cells in the preclinical setting, had any bearing on the activity of immune checkpoint inhibitors alone or in combination with multi-tyrosine kinase inhibitors. The analysis included 54 patients enrolled in a phase 1b trial  of REGONIVO (regorafenib and nivolumab) and a phase 2 trial of LENPEM (lenvatinib and pembrolizumab). With a median follow up of 14 months, there was no significant difference in the efficacy of the above regimens (overall survival, progression-free survival, and objective response rate) between patients with and without liver metastasis. The promising activity of these combinations is continuing with longer follow-up. The above regimens should be investigated further in larger prospective studies irrespective of metastatic sites.

Additional References

1.           Al-Batran SE, Homann N, Pauligk C, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet. 2019;393:1948-1957. Doi: 10.1016/S0140-6736(18)32557-1

2.           Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11-20. Doi:  10.1056/NEJMoa055531

3.           Cunningham D, Starling N, Rao S, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36-46. Doi: 10.1056/NEJMoa073149

4.           Smyth EC, Wotherspoon A, Peckitt C, et al. Mismatch repair deficiency, microsatellite instability, and survival: an exploratory analysis of the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) trial. JAMA Oncol. 2017;3:1197-1203. Doi: 10.1001/jamaoncol.2016.6762

5.           Andre T, Tougeron D, Piessen G, et al. Neoadjuvant nivolumab plus ipilimumab and adjuvant nivolumab in patients (pts) with localized microsatellite instability-high (MSI)/mismatch repair deficient (dMMR) oeso-gastric adenocarcinoma (OGA): the GERCOR NEONIPIGA phase II study. J Clin Oncol. 2022;40:244-244. Doi: 10.1200/JCO.2022.40.4_suppl.244

 

 

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Publications
Topics
Sections
Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Dr Uboha scans the journals so you don't have to!
Dr Uboha scans the journals so you don't have to!

 

Patients with stage II or III gastric cancer are treated with surgical resection and perioperative chemotherapy. Platinum agents have established activity in this disease. Combination chemotherapy FLOT (5-fluorouracil, oxaliplatin, and docetaxel) is now standard perioperative treatment for resectable gastric cancer.1 The study by Slagter and colleagues evaluated whether cisplatin was noninferior to oxaliplatin when used in the treatment of early-stage gastric cancer. Prior to the incorporation of FLOT into standard treatment practice, patients were treated with ECX (epirubicin, cisplatin, and docetaxel), as per the MAGIC trial.2 In the metastatic setting, chemotherapy regimens with either cisplatin or oxaliplatin as a choice of platinum agent have comparable activity against these tumors. Oxaliplatin activity has been shown to be noninferior to cisplatin in the randomized REAL2 trial in metastatic setting.3

The study by Slagter and colleagues is a post hoc analysis of 781 patients with resectable gastric cancer who were enrolled in the CRITICS trial. This analysis demonstrated that chemotherapy regimens containing oxaliplatin and cisplatin had comparable 5-year overall survival rates. Not surprisingly, oxaliplatin was associated with higher neurotoxicity. Based on this analysis, it is likely safe to conclude that, just as in the advanced setting, cisplatin and oxaliplatin have similar activity in early-stage disease.

Mismatch repair protein deficient or microsatellite unstable gastric cancer (MSI-H) represent unique subtypes of gastric cancer, with distinct biologic behaviors and treatment responses. The efficacy of chemotherapy in patients with early-stage MSI-H tumors has been questioned previously. Similar to MSI-H colorectal cancers, the benefit of chemotherapy in resectable MSI-H gastric and esophagogastric junction tumors appears to be less robust than in microsatellite stable (MSS) tumors. In the exploratory analysis of patients with MSI-H tumors enrolled in the perioperative MAGIC trial, patients with MSI-H tumors had better prognosis when treated with surgery alone and potentially experienced detrimental effects from chemotherapy.4 The retrospective analysis by Vos and colleagues adds to the body of knowledge about early-stage MSI-H gastric cancers. They evaluated 535 patients with early-stage disease who were treated with surgery alone or surgery plus perioperative therapy between 2000 and 2018. The overall survival in 82 patients with MSI-H tumors was 20% better than in those with MSS disease. This favorable outcome was seen irrespective of whether chemotherapy was given. Though these results suggest that chemotherapy may not be necessary in the treatment of these tumors and there are emerging data regarding the activity of immune checkpoint inhibitors in this setting, these results should definitely be investigated further in prospective studies.5 However, in the absence of prospective randomized data, it is difficult to recommend deviating from the established standard of care with FLOT, especially for patients undergoing curative intent treatment.

A study by Yukami and colleagues evaluated whether the presence of liver metastasis, which have been shown to be enriched in immunosuppressive cells in the preclinical setting, had any bearing on the activity of immune checkpoint inhibitors alone or in combination with multi-tyrosine kinase inhibitors. The analysis included 54 patients enrolled in a phase 1b trial  of REGONIVO (regorafenib and nivolumab) and a phase 2 trial of LENPEM (lenvatinib and pembrolizumab). With a median follow up of 14 months, there was no significant difference in the efficacy of the above regimens (overall survival, progression-free survival, and objective response rate) between patients with and without liver metastasis. The promising activity of these combinations is continuing with longer follow-up. The above regimens should be investigated further in larger prospective studies irrespective of metastatic sites.

Additional References

1.           Al-Batran SE, Homann N, Pauligk C, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet. 2019;393:1948-1957. Doi: 10.1016/S0140-6736(18)32557-1

2.           Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11-20. Doi:  10.1056/NEJMoa055531

3.           Cunningham D, Starling N, Rao S, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36-46. Doi: 10.1056/NEJMoa073149

4.           Smyth EC, Wotherspoon A, Peckitt C, et al. Mismatch repair deficiency, microsatellite instability, and survival: an exploratory analysis of the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) trial. JAMA Oncol. 2017;3:1197-1203. Doi: 10.1001/jamaoncol.2016.6762

5.           Andre T, Tougeron D, Piessen G, et al. Neoadjuvant nivolumab plus ipilimumab and adjuvant nivolumab in patients (pts) with localized microsatellite instability-high (MSI)/mismatch repair deficient (dMMR) oeso-gastric adenocarcinoma (OGA): the GERCOR NEONIPIGA phase II study. J Clin Oncol. 2022;40:244-244. Doi: 10.1200/JCO.2022.40.4_suppl.244

 

 

 

Patients with stage II or III gastric cancer are treated with surgical resection and perioperative chemotherapy. Platinum agents have established activity in this disease. Combination chemotherapy FLOT (5-fluorouracil, oxaliplatin, and docetaxel) is now standard perioperative treatment for resectable gastric cancer.1 The study by Slagter and colleagues evaluated whether cisplatin was noninferior to oxaliplatin when used in the treatment of early-stage gastric cancer. Prior to the incorporation of FLOT into standard treatment practice, patients were treated with ECX (epirubicin, cisplatin, and docetaxel), as per the MAGIC trial.2 In the metastatic setting, chemotherapy regimens with either cisplatin or oxaliplatin as a choice of platinum agent have comparable activity against these tumors. Oxaliplatin activity has been shown to be noninferior to cisplatin in the randomized REAL2 trial in metastatic setting.3

The study by Slagter and colleagues is a post hoc analysis of 781 patients with resectable gastric cancer who were enrolled in the CRITICS trial. This analysis demonstrated that chemotherapy regimens containing oxaliplatin and cisplatin had comparable 5-year overall survival rates. Not surprisingly, oxaliplatin was associated with higher neurotoxicity. Based on this analysis, it is likely safe to conclude that, just as in the advanced setting, cisplatin and oxaliplatin have similar activity in early-stage disease.

Mismatch repair protein deficient or microsatellite unstable gastric cancer (MSI-H) represent unique subtypes of gastric cancer, with distinct biologic behaviors and treatment responses. The efficacy of chemotherapy in patients with early-stage MSI-H tumors has been questioned previously. Similar to MSI-H colorectal cancers, the benefit of chemotherapy in resectable MSI-H gastric and esophagogastric junction tumors appears to be less robust than in microsatellite stable (MSS) tumors. In the exploratory analysis of patients with MSI-H tumors enrolled in the perioperative MAGIC trial, patients with MSI-H tumors had better prognosis when treated with surgery alone and potentially experienced detrimental effects from chemotherapy.4 The retrospective analysis by Vos and colleagues adds to the body of knowledge about early-stage MSI-H gastric cancers. They evaluated 535 patients with early-stage disease who were treated with surgery alone or surgery plus perioperative therapy between 2000 and 2018. The overall survival in 82 patients with MSI-H tumors was 20% better than in those with MSS disease. This favorable outcome was seen irrespective of whether chemotherapy was given. Though these results suggest that chemotherapy may not be necessary in the treatment of these tumors and there are emerging data regarding the activity of immune checkpoint inhibitors in this setting, these results should definitely be investigated further in prospective studies.5 However, in the absence of prospective randomized data, it is difficult to recommend deviating from the established standard of care with FLOT, especially for patients undergoing curative intent treatment.

A study by Yukami and colleagues evaluated whether the presence of liver metastasis, which have been shown to be enriched in immunosuppressive cells in the preclinical setting, had any bearing on the activity of immune checkpoint inhibitors alone or in combination with multi-tyrosine kinase inhibitors. The analysis included 54 patients enrolled in a phase 1b trial  of REGONIVO (regorafenib and nivolumab) and a phase 2 trial of LENPEM (lenvatinib and pembrolizumab). With a median follow up of 14 months, there was no significant difference in the efficacy of the above regimens (overall survival, progression-free survival, and objective response rate) between patients with and without liver metastasis. The promising activity of these combinations is continuing with longer follow-up. The above regimens should be investigated further in larger prospective studies irrespective of metastatic sites.

Additional References

1.           Al-Batran SE, Homann N, Pauligk C, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet. 2019;393:1948-1957. Doi: 10.1016/S0140-6736(18)32557-1

2.           Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11-20. Doi:  10.1056/NEJMoa055531

3.           Cunningham D, Starling N, Rao S, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36-46. Doi: 10.1056/NEJMoa073149

4.           Smyth EC, Wotherspoon A, Peckitt C, et al. Mismatch repair deficiency, microsatellite instability, and survival: an exploratory analysis of the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) trial. JAMA Oncol. 2017;3:1197-1203. Doi: 10.1001/jamaoncol.2016.6762

5.           Andre T, Tougeron D, Piessen G, et al. Neoadjuvant nivolumab plus ipilimumab and adjuvant nivolumab in patients (pts) with localized microsatellite instability-high (MSI)/mismatch repair deficient (dMMR) oeso-gastric adenocarcinoma (OGA): the GERCOR NEONIPIGA phase II study. J Clin Oncol. 2022;40:244-244. Doi: 10.1200/JCO.2022.40.4_suppl.244

 

 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Gastric Cancer, August 2022
Gate On Date
Sun, 05/01/2022 - 20:00
Un-Gate On Date
Sun, 05/01/2022 - 20:00
Use ProPublica
CFC Schedule Remove Status
Sun, 05/01/2022 - 20:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
323615.9
Activity ID
84692
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Erbitux [ 4139 ]

Commentary: Ramucimumab, Napabucasin, and Pembrolizumab Trial Results, July 2022

Article Type
Changed
Fri, 07/01/2022 - 13:48
Dr Uboha scans the journals so you don't have to!

 

Patients with advanced gastroesophageal adenocarcinoma (GEA) have a poor prognosis. Despite recent improvements in treatments, overall survival (OS) for most of these patients remains < 18 months. Immune checkpoint inhibitors in combination with chemotherapy are now approved for treatment of advanced GEA in the first-line setting (pembrolizumab for esophageal cancers and nivolumab for GEA). These approvals are based on the results of the randomized phase 3 studies KEYNOTE-590 and CheckMate 649.1,2 However, there remains a need to better define patients who are more likely to benefit from these agents.

KEYNOTE-062 was a phase 3 study of pembrolizumab in patients with advanced gastric and gastroesophageal junctionadenocarcinoma.Patients were randomly assignedto receive chemotherapy, chemotherapy plus pembrolizumab, or pembrolizumab alone. This study did not lead to pembrolizumab approval for gastric cancer or GEA, although, consistently with other studies, it suggested that patients whose tumors had higher programmed death-ligand 1 expression were more likely to benefit from immunotherapy treatments.

Lee and colleagues published results of a prespecified exploratory analysis from this study that evaluated the association between tumor mutational burden (TMB) and patient outcomes. Those who had tumors with higher TMB had better outcomes from immunotherapy treatments, but not chemotherapy alone. In patients with TMB ≥10 mut/Mb, pembrolizumab vs chemotherapy significantly improved the objective response rate (ORR; 55.6% vs 41.2%), progression-free survival (PFS; 11.1 vs. 7.0 months; hazard ratio [HR] 0.52) and OS (31.6 vs 13.4 months; HR 0.34). Similarly, pembrolizumab plus chemotherapy vs chemotherapy alone improved ORR (73.3% vs 41.2%), PFS (31.1 vs 7.0 months; HR 0.62), and OS (31.1 vs 13.4 months; HR 0.54). However, after patients with microsatellite instability–high (MSI-H) tumors, who represented 44% of patients (22 of 50), with TMB 10 mut/Mb were removed from the analysis, the positive association between TMB and immunotherapy effect on OS and PFS was no longer significant. The small number of patients with high TMB limits the interpretation of these exploratory data. However, the results suggest that TMB is unlikely to emerge as a significant clinically relevant biomarker in patients with microsatellite-stable upper gastrointestinal cancers.

Ultimately, novel therapeutic strategies are needed to improve outcomes. The phase 3 BRIGHTER study reported by Shah and colleagues evaluated the activity of napabucasin, a generator of reactive oxygen species, in advanced GEA. This was a phase 3 study that enrolled 714 patients whose tumors progressed on one prior line of therapy. Patients were randomly assigned in a 1:1 fashion to receive eitherpaclitaxel plus napabucasin or paclitaxel plus placebo. The primary endpoint was OS. Patient characteristics were well balanced between the treatment arms. At the preplanned interim analysis, there was no difference in OS between the two treatment arms (6.97 months with napabucasin vs 7.29 months with placebo; P =.5699).As such, the study was terminated before enrollment completion because it was deemed futile to continue. There were no new safety signals. It is important to note that since the study was launched, ramucirumab and paclitaxel became the new standard treatment in the second-line setting per the phase 3 RAINBOW study.As such, this combination regimen should be offered to patients who are candidates for systemic treatment in the second-line setting and who have no contraindications to these agents.

To further expand on the use of ramucirumab in GEA, the phase 2 HGCSG1603 study evaluated ramucimumab in combination with irinotecan in the second-line setting. This single-arm phase 2 study conducted in Japan enrolled 35 patients with GEA who progressed on a first-line regimen. The primary endpoint was PFS rate at 6 months. This study demonstrated a 26.5% PFS rate at 6 months. Median PFS was 4.2 months, and OS was 9.6 months. Although the study did not meet its pre-specified primary endpoint of a 39% PFS rate at 6 months, the secondary endpoints of OS and PFS were similar to historical references from the RAINBOW study. These results suggest that irinotecan and ramucirumab is an active combination in GEA and could be offered to patients who are not candidates for paclitaxel, for example those with significant neuropathy. The benefits of an irinotecan chemotherapy backbone in second-line therapy has been previously demonstrated in the phase 2 RAMIRIS study, which found clinically meaningful activity of a leucovorin calcium (calcium folinate), 5-fluorouracil, and irinotecan (FOLFIRI) and ramucirumab combination, as well as a decreased benefit from paclitaxel in patients with prior taxane exposure.5 In summary, ramucirumab in combination with irinotecan-containing chemotherapy (either as a single agent or as part of FOLFIRI) is areasonable second-line treatment option for patients with advanced GEA, and this is already included in National Comprehensive Cancer Network guidelines.

 

Additional References

              1.           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. 2021;398:27-40. Doi: 10.1016/S0140-6736(21)00797-2

              2.           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. 2021;398:759-771. Doi: 10.1016/S0140-6736(21)01234-4

              3.           Shitara K, Van Cutsem E, Bang YJ, et al. Efficacy and safety of pembrolizumab or pembrolizumab plus chemotherapy vs chemotherapy alone for patients with first-line, advanced gastric cancer: The KEYNOTE-062 phase 3 randomized clinical trial. JAMA Oncol. 2020;6:1571-1580. Doi: 10.1001/jamaoncol.2020.3370

              4.           Wilke H, Van Cutsem E, Cheul Oh S, et al. RAINBOW: A global, phase 3, randomized, double-blind study of ramucirumab plus paclitaxel versus placebo plus paclitaxel in the treatment of metastatic gastric adenocarcinoma following disease progression on first-line platinum- and fluoropyrimidine-containing combination therapy: Results of a multiple Cox regression analysis adjusting for prognostic factors. J Clin Oncol. 2014;32(15 Suppl):4076. Doi: 10.1200/jco.2014.32.15_suppl.4076

5.           Klempner SJ, Maron SB, Chase L, et al. Initial report of second-line FOLFIRi in combination with ramucirumab in advanced gastroesophageal adenocarcinomas: A multi-institutional retrospective analysis. Oncologist. 2019:24:475-482. Doi: 10.1634/theoncologist.2018-0602

Dr. Nataliya Uboha, MD

 

 

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Publications
Topics
Sections
Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Dr Uboha scans the journals so you don't have to!
Dr Uboha scans the journals so you don't have to!

 

Patients with advanced gastroesophageal adenocarcinoma (GEA) have a poor prognosis. Despite recent improvements in treatments, overall survival (OS) for most of these patients remains < 18 months. Immune checkpoint inhibitors in combination with chemotherapy are now approved for treatment of advanced GEA in the first-line setting (pembrolizumab for esophageal cancers and nivolumab for GEA). These approvals are based on the results of the randomized phase 3 studies KEYNOTE-590 and CheckMate 649.1,2 However, there remains a need to better define patients who are more likely to benefit from these agents.

KEYNOTE-062 was a phase 3 study of pembrolizumab in patients with advanced gastric and gastroesophageal junctionadenocarcinoma.Patients were randomly assignedto receive chemotherapy, chemotherapy plus pembrolizumab, or pembrolizumab alone. This study did not lead to pembrolizumab approval for gastric cancer or GEA, although, consistently with other studies, it suggested that patients whose tumors had higher programmed death-ligand 1 expression were more likely to benefit from immunotherapy treatments.

Lee and colleagues published results of a prespecified exploratory analysis from this study that evaluated the association between tumor mutational burden (TMB) and patient outcomes. Those who had tumors with higher TMB had better outcomes from immunotherapy treatments, but not chemotherapy alone. In patients with TMB ≥10 mut/Mb, pembrolizumab vs chemotherapy significantly improved the objective response rate (ORR; 55.6% vs 41.2%), progression-free survival (PFS; 11.1 vs. 7.0 months; hazard ratio [HR] 0.52) and OS (31.6 vs 13.4 months; HR 0.34). Similarly, pembrolizumab plus chemotherapy vs chemotherapy alone improved ORR (73.3% vs 41.2%), PFS (31.1 vs 7.0 months; HR 0.62), and OS (31.1 vs 13.4 months; HR 0.54). However, after patients with microsatellite instability–high (MSI-H) tumors, who represented 44% of patients (22 of 50), with TMB 10 mut/Mb were removed from the analysis, the positive association between TMB and immunotherapy effect on OS and PFS was no longer significant. The small number of patients with high TMB limits the interpretation of these exploratory data. However, the results suggest that TMB is unlikely to emerge as a significant clinically relevant biomarker in patients with microsatellite-stable upper gastrointestinal cancers.

Ultimately, novel therapeutic strategies are needed to improve outcomes. The phase 3 BRIGHTER study reported by Shah and colleagues evaluated the activity of napabucasin, a generator of reactive oxygen species, in advanced GEA. This was a phase 3 study that enrolled 714 patients whose tumors progressed on one prior line of therapy. Patients were randomly assigned in a 1:1 fashion to receive eitherpaclitaxel plus napabucasin or paclitaxel plus placebo. The primary endpoint was OS. Patient characteristics were well balanced between the treatment arms. At the preplanned interim analysis, there was no difference in OS between the two treatment arms (6.97 months with napabucasin vs 7.29 months with placebo; P =.5699).As such, the study was terminated before enrollment completion because it was deemed futile to continue. There were no new safety signals. It is important to note that since the study was launched, ramucirumab and paclitaxel became the new standard treatment in the second-line setting per the phase 3 RAINBOW study.As such, this combination regimen should be offered to patients who are candidates for systemic treatment in the second-line setting and who have no contraindications to these agents.

To further expand on the use of ramucirumab in GEA, the phase 2 HGCSG1603 study evaluated ramucimumab in combination with irinotecan in the second-line setting. This single-arm phase 2 study conducted in Japan enrolled 35 patients with GEA who progressed on a first-line regimen. The primary endpoint was PFS rate at 6 months. This study demonstrated a 26.5% PFS rate at 6 months. Median PFS was 4.2 months, and OS was 9.6 months. Although the study did not meet its pre-specified primary endpoint of a 39% PFS rate at 6 months, the secondary endpoints of OS and PFS were similar to historical references from the RAINBOW study. These results suggest that irinotecan and ramucirumab is an active combination in GEA and could be offered to patients who are not candidates for paclitaxel, for example those with significant neuropathy. The benefits of an irinotecan chemotherapy backbone in second-line therapy has been previously demonstrated in the phase 2 RAMIRIS study, which found clinically meaningful activity of a leucovorin calcium (calcium folinate), 5-fluorouracil, and irinotecan (FOLFIRI) and ramucirumab combination, as well as a decreased benefit from paclitaxel in patients with prior taxane exposure.5 In summary, ramucirumab in combination with irinotecan-containing chemotherapy (either as a single agent or as part of FOLFIRI) is areasonable second-line treatment option for patients with advanced GEA, and this is already included in National Comprehensive Cancer Network guidelines.

 

Additional References

              1.           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. 2021;398:27-40. Doi: 10.1016/S0140-6736(21)00797-2

              2.           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. 2021;398:759-771. Doi: 10.1016/S0140-6736(21)01234-4

              3.           Shitara K, Van Cutsem E, Bang YJ, et al. Efficacy and safety of pembrolizumab or pembrolizumab plus chemotherapy vs chemotherapy alone for patients with first-line, advanced gastric cancer: The KEYNOTE-062 phase 3 randomized clinical trial. JAMA Oncol. 2020;6:1571-1580. Doi: 10.1001/jamaoncol.2020.3370

              4.           Wilke H, Van Cutsem E, Cheul Oh S, et al. RAINBOW: A global, phase 3, randomized, double-blind study of ramucirumab plus paclitaxel versus placebo plus paclitaxel in the treatment of metastatic gastric adenocarcinoma following disease progression on first-line platinum- and fluoropyrimidine-containing combination therapy: Results of a multiple Cox regression analysis adjusting for prognostic factors. J Clin Oncol. 2014;32(15 Suppl):4076. Doi: 10.1200/jco.2014.32.15_suppl.4076

5.           Klempner SJ, Maron SB, Chase L, et al. Initial report of second-line FOLFIRi in combination with ramucirumab in advanced gastroesophageal adenocarcinomas: A multi-institutional retrospective analysis. Oncologist. 2019:24:475-482. Doi: 10.1634/theoncologist.2018-0602

Dr. Nataliya Uboha, MD

 

 

 

Patients with advanced gastroesophageal adenocarcinoma (GEA) have a poor prognosis. Despite recent improvements in treatments, overall survival (OS) for most of these patients remains < 18 months. Immune checkpoint inhibitors in combination with chemotherapy are now approved for treatment of advanced GEA in the first-line setting (pembrolizumab for esophageal cancers and nivolumab for GEA). These approvals are based on the results of the randomized phase 3 studies KEYNOTE-590 and CheckMate 649.1,2 However, there remains a need to better define patients who are more likely to benefit from these agents.

KEYNOTE-062 was a phase 3 study of pembrolizumab in patients with advanced gastric and gastroesophageal junctionadenocarcinoma.Patients were randomly assignedto receive chemotherapy, chemotherapy plus pembrolizumab, or pembrolizumab alone. This study did not lead to pembrolizumab approval for gastric cancer or GEA, although, consistently with other studies, it suggested that patients whose tumors had higher programmed death-ligand 1 expression were more likely to benefit from immunotherapy treatments.

Lee and colleagues published results of a prespecified exploratory analysis from this study that evaluated the association between tumor mutational burden (TMB) and patient outcomes. Those who had tumors with higher TMB had better outcomes from immunotherapy treatments, but not chemotherapy alone. In patients with TMB ≥10 mut/Mb, pembrolizumab vs chemotherapy significantly improved the objective response rate (ORR; 55.6% vs 41.2%), progression-free survival (PFS; 11.1 vs. 7.0 months; hazard ratio [HR] 0.52) and OS (31.6 vs 13.4 months; HR 0.34). Similarly, pembrolizumab plus chemotherapy vs chemotherapy alone improved ORR (73.3% vs 41.2%), PFS (31.1 vs 7.0 months; HR 0.62), and OS (31.1 vs 13.4 months; HR 0.54). However, after patients with microsatellite instability–high (MSI-H) tumors, who represented 44% of patients (22 of 50), with TMB 10 mut/Mb were removed from the analysis, the positive association between TMB and immunotherapy effect on OS and PFS was no longer significant. The small number of patients with high TMB limits the interpretation of these exploratory data. However, the results suggest that TMB is unlikely to emerge as a significant clinically relevant biomarker in patients with microsatellite-stable upper gastrointestinal cancers.

Ultimately, novel therapeutic strategies are needed to improve outcomes. The phase 3 BRIGHTER study reported by Shah and colleagues evaluated the activity of napabucasin, a generator of reactive oxygen species, in advanced GEA. This was a phase 3 study that enrolled 714 patients whose tumors progressed on one prior line of therapy. Patients were randomly assigned in a 1:1 fashion to receive eitherpaclitaxel plus napabucasin or paclitaxel plus placebo. The primary endpoint was OS. Patient characteristics were well balanced between the treatment arms. At the preplanned interim analysis, there was no difference in OS between the two treatment arms (6.97 months with napabucasin vs 7.29 months with placebo; P =.5699).As such, the study was terminated before enrollment completion because it was deemed futile to continue. There were no new safety signals. It is important to note that since the study was launched, ramucirumab and paclitaxel became the new standard treatment in the second-line setting per the phase 3 RAINBOW study.As such, this combination regimen should be offered to patients who are candidates for systemic treatment in the second-line setting and who have no contraindications to these agents.

To further expand on the use of ramucirumab in GEA, the phase 2 HGCSG1603 study evaluated ramucimumab in combination with irinotecan in the second-line setting. This single-arm phase 2 study conducted in Japan enrolled 35 patients with GEA who progressed on a first-line regimen. The primary endpoint was PFS rate at 6 months. This study demonstrated a 26.5% PFS rate at 6 months. Median PFS was 4.2 months, and OS was 9.6 months. Although the study did not meet its pre-specified primary endpoint of a 39% PFS rate at 6 months, the secondary endpoints of OS and PFS were similar to historical references from the RAINBOW study. These results suggest that irinotecan and ramucirumab is an active combination in GEA and could be offered to patients who are not candidates for paclitaxel, for example those with significant neuropathy. The benefits of an irinotecan chemotherapy backbone in second-line therapy has been previously demonstrated in the phase 2 RAMIRIS study, which found clinically meaningful activity of a leucovorin calcium (calcium folinate), 5-fluorouracil, and irinotecan (FOLFIRI) and ramucirumab combination, as well as a decreased benefit from paclitaxel in patients with prior taxane exposure.5 In summary, ramucirumab in combination with irinotecan-containing chemotherapy (either as a single agent or as part of FOLFIRI) is areasonable second-line treatment option for patients with advanced GEA, and this is already included in National Comprehensive Cancer Network guidelines.

 

Additional References

              1.           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. 2021;398:27-40. Doi: 10.1016/S0140-6736(21)00797-2

              2.           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. 2021;398:759-771. Doi: 10.1016/S0140-6736(21)01234-4

              3.           Shitara K, Van Cutsem E, Bang YJ, et al. Efficacy and safety of pembrolizumab or pembrolizumab plus chemotherapy vs chemotherapy alone for patients with first-line, advanced gastric cancer: The KEYNOTE-062 phase 3 randomized clinical trial. JAMA Oncol. 2020;6:1571-1580. Doi: 10.1001/jamaoncol.2020.3370

              4.           Wilke H, Van Cutsem E, Cheul Oh S, et al. RAINBOW: A global, phase 3, randomized, double-blind study of ramucirumab plus paclitaxel versus placebo plus paclitaxel in the treatment of metastatic gastric adenocarcinoma following disease progression on first-line platinum- and fluoropyrimidine-containing combination therapy: Results of a multiple Cox regression analysis adjusting for prognostic factors. J Clin Oncol. 2014;32(15 Suppl):4076. Doi: 10.1200/jco.2014.32.15_suppl.4076

5.           Klempner SJ, Maron SB, Chase L, et al. Initial report of second-line FOLFIRi in combination with ramucirumab in advanced gastroesophageal adenocarcinomas: A multi-institutional retrospective analysis. Oncologist. 2019:24:475-482. Doi: 10.1634/theoncologist.2018-0602

Dr. Nataliya Uboha, MD

 

 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan; Gastric Cancer, July 2022
Gate On Date
Sun, 05/01/2022 - 20:00
Un-Gate On Date
Sun, 05/01/2022 - 20:00
Use ProPublica
CFC Schedule Remove Status
Sun, 05/01/2022 - 20:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
323615.9
Activity ID
84692
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Erbitux [ 4139 ]

Commentary: Gastric Cancer Chemotherapy in Select Populations, June 2022

Article Type
Changed
Thu, 06/09/2022 - 11:48
Dr Uboha scans the journals so you don't have to!

 

Dr. Nataliya Uboha, MD

Chemotherapy plays a critical role in the management of patients with advanced gastroesophageal adenocarcinoma, and a fluoropyrimidine and platinum doublet (either oxaliplatin or cisplatin) is the standard regimen used. Chemotherapy is associated with toxicity, which is particularly concerning in frail and older adult patients.1 A study by Chinen and colleagues specifically looked at the use of platinum chemotherapy agents in the older adult patient population. This retrospective cohort study analyzed survival outcomes and granulocyte colony–stimulating factor (G-CSF) use in 242 patients with advanced gastric cancer who were at least 70 years old and who were treated with either an oxaliplatin- or cisplatin-containing regimen. After propensity score weighting, the study demonstrated that treatment with these agents resulted in similar overall survival, but G-CSF use was more frequent with oxaliplatin use. These results are in line with previous data regarding these agents. In a prospective noninferiority study with a two-by-two design (REAL-2), an oxaliplatin-containing regimen had similar activity to a cisplatin-containing regimen.2 However, oxaliplatin use was associated with less neutropenia. Going forward, the use of oxaliplatin should be preferred over cisplatin in older adult patients given its more favorable toxicity profile.

A study by Sotelo and colleagues looked at the presence of gastric preneoplastic lesions in the first-degree relatives of patients with gastric cancer. In this cross-sectional study conducted in Chile, endoscopic evaluation was performed in 110 people eligible for evaluation. Among the participants, 95 cases (86.4%) of preneoplastic lesions were identified, most commonly atrophic gastritis (86.4%) and intestinal metaplasia (82.7%). There was no association with sex, age, or Helicobacter pylori infection. The high rates of these lesions in the study are probably reflective of a high prevalence of gastric cancer in this geographic area. Although the data interpretation is limited by the small study size, these results suggest that endoscopic surveillance of first-degree relatives is warranted in areas of high gastric cancer incidence, and that optimal surveillance protocols, as well as management of these precancerous conditions, should be defined further.

The role of hyperthermic intraperitoneal chemotherapy (HIPEC) has been controversial in the management of gastric cancer. Thus far, there has not been a definitive study demonstrating a positive impact of HIPEC on survival in patients with this disease. A study by Lee and colleagues looked at the role of prophylactic HIPEC in patients with clinical stage T4 gastric cancer who do not have evidence of distant metastasis. Retrospective analysis included 132 patients with clinical stage T4 gastric cancer who underwent gastrectomy and D2 lymphadenectomy. After propensity score matching to reduce selection bias, the outcomes of 35 patients per cohort (gastrectomy vs gastrectomy plus prophylactic HIPEC) were analyzed. The two groups were well matched in regard to histology, pathologic T and N stage, perioperative therapy, and the type of resection. There was no difference in the incidence of postoperative complications. While the rate of peritoneal recurrences was lower in the HIPEC cohort, there was no difference in the rate of distant metastasis between the two groups. In terms of survival outcomes, the study demonstrated that disease-free survival and overall survival were improved with prophylactic HIPEC. Although firm conclusions cannot be drawn from this small retrospective study, these results suggest that it may be worth further investigating the role of prophylactic HIPEC in a subset of patients with high-risk early-stage disease.

Additional References

1. Hwang IG, Ji JH, Kang JH, et al. A multi-center, open-label, randomized phase III trial of first-line chemotherapy with capecitabine monotherapy versus capecitabine plus oxaliplatin in elderly patients with advanced gastric cancer. J Geriatr Oncol. 2017;8(3):170-175. Doi: 10.1016/j.jgo.2017.01.002

2. Cunningham D, Starling N, Rao S, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36-46. Doi:  10.1056/NEJMoa073149

 

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Publications
Topics
Sections
Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Author and Disclosure Information

Faculty Leader for Cancer Therapy Discovery & Development/Phase I Program, Associate Professor, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, Madison, WI

Dr Uboha scans the journals so you don't have to!
Dr Uboha scans the journals so you don't have to!

 

Dr. Nataliya Uboha, MD

Chemotherapy plays a critical role in the management of patients with advanced gastroesophageal adenocarcinoma, and a fluoropyrimidine and platinum doublet (either oxaliplatin or cisplatin) is the standard regimen used. Chemotherapy is associated with toxicity, which is particularly concerning in frail and older adult patients.1 A study by Chinen and colleagues specifically looked at the use of platinum chemotherapy agents in the older adult patient population. This retrospective cohort study analyzed survival outcomes and granulocyte colony–stimulating factor (G-CSF) use in 242 patients with advanced gastric cancer who were at least 70 years old and who were treated with either an oxaliplatin- or cisplatin-containing regimen. After propensity score weighting, the study demonstrated that treatment with these agents resulted in similar overall survival, but G-CSF use was more frequent with oxaliplatin use. These results are in line with previous data regarding these agents. In a prospective noninferiority study with a two-by-two design (REAL-2), an oxaliplatin-containing regimen had similar activity to a cisplatin-containing regimen.2 However, oxaliplatin use was associated with less neutropenia. Going forward, the use of oxaliplatin should be preferred over cisplatin in older adult patients given its more favorable toxicity profile.

A study by Sotelo and colleagues looked at the presence of gastric preneoplastic lesions in the first-degree relatives of patients with gastric cancer. In this cross-sectional study conducted in Chile, endoscopic evaluation was performed in 110 people eligible for evaluation. Among the participants, 95 cases (86.4%) of preneoplastic lesions were identified, most commonly atrophic gastritis (86.4%) and intestinal metaplasia (82.7%). There was no association with sex, age, or Helicobacter pylori infection. The high rates of these lesions in the study are probably reflective of a high prevalence of gastric cancer in this geographic area. Although the data interpretation is limited by the small study size, these results suggest that endoscopic surveillance of first-degree relatives is warranted in areas of high gastric cancer incidence, and that optimal surveillance protocols, as well as management of these precancerous conditions, should be defined further.

The role of hyperthermic intraperitoneal chemotherapy (HIPEC) has been controversial in the management of gastric cancer. Thus far, there has not been a definitive study demonstrating a positive impact of HIPEC on survival in patients with this disease. A study by Lee and colleagues looked at the role of prophylactic HIPEC in patients with clinical stage T4 gastric cancer who do not have evidence of distant metastasis. Retrospective analysis included 132 patients with clinical stage T4 gastric cancer who underwent gastrectomy and D2 lymphadenectomy. After propensity score matching to reduce selection bias, the outcomes of 35 patients per cohort (gastrectomy vs gastrectomy plus prophylactic HIPEC) were analyzed. The two groups were well matched in regard to histology, pathologic T and N stage, perioperative therapy, and the type of resection. There was no difference in the incidence of postoperative complications. While the rate of peritoneal recurrences was lower in the HIPEC cohort, there was no difference in the rate of distant metastasis between the two groups. In terms of survival outcomes, the study demonstrated that disease-free survival and overall survival were improved with prophylactic HIPEC. Although firm conclusions cannot be drawn from this small retrospective study, these results suggest that it may be worth further investigating the role of prophylactic HIPEC in a subset of patients with high-risk early-stage disease.

Additional References

1. Hwang IG, Ji JH, Kang JH, et al. A multi-center, open-label, randomized phase III trial of first-line chemotherapy with capecitabine monotherapy versus capecitabine plus oxaliplatin in elderly patients with advanced gastric cancer. J Geriatr Oncol. 2017;8(3):170-175. Doi: 10.1016/j.jgo.2017.01.002

2. Cunningham D, Starling N, Rao S, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36-46. Doi:  10.1056/NEJMoa073149

 

 

Dr. Nataliya Uboha, MD

Chemotherapy plays a critical role in the management of patients with advanced gastroesophageal adenocarcinoma, and a fluoropyrimidine and platinum doublet (either oxaliplatin or cisplatin) is the standard regimen used. Chemotherapy is associated with toxicity, which is particularly concerning in frail and older adult patients.1 A study by Chinen and colleagues specifically looked at the use of platinum chemotherapy agents in the older adult patient population. This retrospective cohort study analyzed survival outcomes and granulocyte colony–stimulating factor (G-CSF) use in 242 patients with advanced gastric cancer who were at least 70 years old and who were treated with either an oxaliplatin- or cisplatin-containing regimen. After propensity score weighting, the study demonstrated that treatment with these agents resulted in similar overall survival, but G-CSF use was more frequent with oxaliplatin use. These results are in line with previous data regarding these agents. In a prospective noninferiority study with a two-by-two design (REAL-2), an oxaliplatin-containing regimen had similar activity to a cisplatin-containing regimen.2 However, oxaliplatin use was associated with less neutropenia. Going forward, the use of oxaliplatin should be preferred over cisplatin in older adult patients given its more favorable toxicity profile.

A study by Sotelo and colleagues looked at the presence of gastric preneoplastic lesions in the first-degree relatives of patients with gastric cancer. In this cross-sectional study conducted in Chile, endoscopic evaluation was performed in 110 people eligible for evaluation. Among the participants, 95 cases (86.4%) of preneoplastic lesions were identified, most commonly atrophic gastritis (86.4%) and intestinal metaplasia (82.7%). There was no association with sex, age, or Helicobacter pylori infection. The high rates of these lesions in the study are probably reflective of a high prevalence of gastric cancer in this geographic area. Although the data interpretation is limited by the small study size, these results suggest that endoscopic surveillance of first-degree relatives is warranted in areas of high gastric cancer incidence, and that optimal surveillance protocols, as well as management of these precancerous conditions, should be defined further.

The role of hyperthermic intraperitoneal chemotherapy (HIPEC) has been controversial in the management of gastric cancer. Thus far, there has not been a definitive study demonstrating a positive impact of HIPEC on survival in patients with this disease. A study by Lee and colleagues looked at the role of prophylactic HIPEC in patients with clinical stage T4 gastric cancer who do not have evidence of distant metastasis. Retrospective analysis included 132 patients with clinical stage T4 gastric cancer who underwent gastrectomy and D2 lymphadenectomy. After propensity score matching to reduce selection bias, the outcomes of 35 patients per cohort (gastrectomy vs gastrectomy plus prophylactic HIPEC) were analyzed. The two groups were well matched in regard to histology, pathologic T and N stage, perioperative therapy, and the type of resection. There was no difference in the incidence of postoperative complications. While the rate of peritoneal recurrences was lower in the HIPEC cohort, there was no difference in the rate of distant metastasis between the two groups. In terms of survival outcomes, the study demonstrated that disease-free survival and overall survival were improved with prophylactic HIPEC. Although firm conclusions cannot be drawn from this small retrospective study, these results suggest that it may be worth further investigating the role of prophylactic HIPEC in a subset of patients with high-risk early-stage disease.

Additional References

1. Hwang IG, Ji JH, Kang JH, et al. A multi-center, open-label, randomized phase III trial of first-line chemotherapy with capecitabine monotherapy versus capecitabine plus oxaliplatin in elderly patients with advanced gastric cancer. J Geriatr Oncol. 2017;8(3):170-175. Doi: 10.1016/j.jgo.2017.01.002

2. Cunningham D, Starling N, Rao S, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36-46. Doi:  10.1056/NEJMoa073149

 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Gastric Cancer June 2022
Gate On Date
Sun, 05/01/2022 - 20:00
Un-Gate On Date
Sun, 05/01/2022 - 20:00
Use ProPublica
CFC Schedule Remove Status
Sun, 05/01/2022 - 20:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
323615.9
Activity ID
84692
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Erbitux [ 4139 ]