Skip to main content

ORIGINAL RESEARCH article

Front. Oncol., 07 March 2022
Sec. Gastrointestinal Cancers: Gastric and Esophageal Cancers

Interest of the Addition of Taxanes to Standard Treatment in First-Line Advanced HER2 Positive Gastroesophageal Adenocarcinoma in Selective Patients

Emeline Orillard,*Emeline Orillard1,2*Julie Henriques,Julie Henriques2,3Dewi Vernerey,Dewi Vernerey2,3Hamadi AlmotlakHamadi Almotlak1Fabien CalcagnoFabien Calcagno1Francine FeinFrancine Fein4Serge FrattSerge Fratté5Marine Jary,Marine Jary1,2Elodie KlajerElodie Klajer1Angelique Vienot,Angelique Vienot1,2Christophe Borg,Christophe Borg1,2Stefano Kim,,Stefano Kim1,2,6
  • 1Department of Oncology, University Hospital of Besançon, Besançon, France
  • 2Bourgogne Franche-Comté University, INSERM, Établissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
  • 3Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France
  • 4Department of Gastroenterology, University Hospital of Besançon, Besançon, France
  • 5Department of Gastroenterology, Centre Hospitalier Régional, Belfort, France
  • 6Department of Oncology and Radiotherapy, Hôpital Nord-Franche Comté, Montbéliard, France

Background: Studies have reported a beneficial role of the addition of trastuzumab to platin-5-FU based chemotherapy in first-line advanced HER2 positive gastroesophageal adenocarcinoma (GEA). However, the effect of taxanes combined with platin-5FU + trastuzumab (PFT) is understudied.

Methods: We performed a retrospective cohort study to evaluate the interest of taxanes among HER2-positive advanced GEA patients treated with PFT. We enrolled HER2-positive advanced GEA patients who underwent treatment between January 2009 to March 2021 in seven hospitals centers in France, treated with PFT alone (S group) or with taxanes + PFT regimen (T group). The primary outcome was progression-free survival (PFS). Also, overall survival (OS), response rate, conversion surgery rate, and safety were evaluated.

Results: Overall, 65 patients received PFT-based therapy, 24 patients in the T group, and 41 patients in the S group. To avoid the selection bias, only those patients presenting an ECOG-PS of 0-1 and synchronous metastasis (21 patients in the T group and 19 patients in the S group) were included for analysis. The median PFS was 9.3 months (95%CI 7.0 to 17.2) in the T group and 5.9 months (95%CI 3.7 to 9.6) in the S group (log-rank p=0.038). Treatment by taxanes was significantly associated with a better PFS in univariate (HR 0.49; 95%CI 0.25 to 0.98, p=0.042) and multivariate Cox regression analysis (HR 0.44; 95%CI 0.21 to 0.94, p=0.033), and IPTW method (HR 0.56; 95% CI 0.34 to 0.91, p=0.019). OS was prolonged (19.0 months (95%CI 7.8 to 45.2) vs 13.0 months (95%CI 5.5 to 14.8), log-rank p=0.033) in favor of the T group. Treatment by taxanes was significantly associated with a better OS in univariate Cox regression analysis (HR 0.49; 95%CI 0.21 to 0.96, p=0.038) and IPTW method (HR 0.49; 95% CI 0.29 to 0.84, p=0.009). The response rate was higher in the T group, with conversion surgery in five patients. No treatment-related death was observed in both groups.

Conclusions: Given the improvement in PFS and OS, the addition of taxanes to standard chemotherapy could be considered as a promising treatment for selected HER2-positive advanced GEA patients, with PS 0-1 and synchronous metastasis (NCT04920747).

Introduction

Fluoropyrimidine and platinum-based chemotherapy, in association with trastuzumab, an anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody, is the standard of care in HER2-positive advanced gastroesophageal adenocarcinoma (GEA) patients, based on the results of the ToGA study published in 2010 (1). Then, all additional efforts to improve anti-HER2 inhibition in first- and second-line, failed to prolong survival. In the second line, trastuzumab maintenance beyond progression, in combination with paclitaxel, failed to improve PFS compared to paclitaxel alone (2). Besides, lapatinib (a tyrosine kinase inhibitor of epidermal growth factor receptor HER1 and HER2) plus paclitaxel (3), or trastuzumab-emtansine (TDM-1), an antibody-drug conjugate (4), also failed to improve survival compared to paclitaxel alone in phase III trials. In first-line, the JACOB trial group has proposed to intensity anti-HER2 blockade with the addition of pertuzumab to trastuzumab and standard chemotherapy (5), and the HELOISE trial group proposed to increase the dose of trastuzumab (6). Unfortunately, both phase III trials were negative, and no survival improvement was seen in HER2-positive GEA patients for 10 years. Recently, in an open-label randomized phase II trial, a new antibody-drug conjugate trastuzumab-deruxtecan showed encouraging results in third- or later line with a significant improvement in objective response rate (51% vs 14%; p<0.001) and overall survival (HR, 0.59; p=0.01) (7). A confirmatory phase III trial is ongoing (DESTINY-Gastric04).

These negative results are contrary to what we have seen in HER2-positive breast cancer, for which the intensification of HER2 inhibition in first-line, as well as, the continuous exposition to an anti-HER2 therapy remains a standard of care in metastatic HER2-positive breast cancer patients setting. Several issues had been highlighted in GEA as possible explanations, like the tumor heterogeneity (8) or the presence of molecular mechanisms of resistance to anti-HER2 therapy, such as changes in the tridimensional receptor structures, the co-expression of other transmembrane receptors, or the activation of downstream signaling effectors (9). Moreover, in the case of secondary resistance to anti-HER2 inhibitor, the loss of HER2 expression has been described (2, 10). In fact, among those patients with new tumor biopsy at progression under trastuzumab in first-line, about 60-70% of patients have lost the HER2 expression in their tumors.

To date, the prognosis of HER2-positive GEA patients remains bleak, with an estimated median overall survival of 14 months after the introduction of the first-line therapy (1, 5). Therefore, there is still an unmet need in this high-risk population. Considering the lack of efficacy of anti-HER2-based intensification therapy, it seems reasonable to optimize the first-line treatment of these patients with a synergic chemotherapy. Our hypothesis is presented in Figure 1. The rationale to combine taxanes to the standard regimen in HER2-positive advanced GEA is based on I) their demonstrated efficacy in the first line in HER2-negative GEA patients, and II) the synergic effect in preclinical and clinical settings between trastuzumab and taxanes-based chemotherapy in HER2-positive tumors. In a phase III trial, Van Cutsem et al. (11) have shown in these patients that the addition of docetaxel to cisplatin and 5-Fluorouracil (standard DCF) as the first-line therapy significantly improved time to progression, overall survival, and response rate. More recently, a randomized phase II study has compared dose-modified docetaxel, cisplatin, and 5-FU (mDCF) to standard DCF regimen in previously untreated patients with advanced GEA (12). Toxicity was significantly decreased with mDCF with improved efficacy and was recommended as a new option for first-line therapy (13). In the preclinical model of breast cancer cell lines, the efficacy of trastuzumab in combination with different types of chemotherapy was evaluated. It was observed synergistic interaction between the use of trastuzumab and docetaxel, and an additive interaction between trastuzumab and paclitaxel (1416), including an enhancement of trastuzumab-mediated antibody-dependent cellular cytotoxicity (ADCC) on tumor cells by taxanes through NKG2D-mediated NK cell recognition (17). In HER2-positive GEA patients in first-line, Al Batran et al. have shown for the first time in 2012 a report of 3 cases of patients that adding trastuzumab to a docetaxel-based, triple-drug chemotherapy combination is feasible and highly active (18). Since then, several single-arm small phase II trials evaluated the association of taxanes, platin salt, 5-FU, and trastuzumab regimens (1921). In these studies, including 15 to 26 patients, this combination was feasible, and the mPFS was 9 to 13 months. One study had mPFS not reached during the median follow-up time of 18.3 months. The overall response rate was 60% to 93%.

FIGURE 1
www.frontiersin.org

Figure 1 The hypothesis to combine taxanes to standard treatment in first-line advanced HER2 positive gastroesophageal adenocarcinoma.

Thus, to evaluate the potential interest of the addition of taxanes to trastuzumab and standard chemotherapy for HER2-positive advanced GEA in the first line, we performed a retrospective multicenter TanDHER study to compare taxanes, platin, 5-FU, and trastuzumab (TPFT) regimen with standard chemotherapy and trastuzumab.

Patients and Methods

Patient Selection

TanDHER (NCT04920747) was a retrospective study undertaken in 7 centers in the Franche-Comte region in France. Patients treated between January 2009 and Mars 2021 were included. Men or women older than 17 years of age were eligible for inclusion if they had histologically confirmed (by tumor biopsy or on a surgical specimen and confirmed by the pathologists of each center) non-resectable locally advanced, recurrent, or metastatic adenocarcinoma of the stomach or gastro-esophageal junction; tumor samples scored as 3+ on immunohistochemistry (IHC) or a FISH (Fluorescence in situ Hybridization) positive tumor (HER2:CEP17 ratio ≥2) with a 2+ IHC score, based on This criterion is based on the guideline from the College of American Pathologists, American Society for Clinical Pathology, and American Society of Clinical Oncology regarding HER2 assessment in gastroesophageal adenocarcinoma (22, 23); measurable disease; treated by trastuzumab, platin (cisplatin or oxaliplatin), fluoropyrimidine (5-FU or capecitabine) +/- taxanes (docetaxel or paclitaxel), as first-line therapy for advanced gastric cancer. Major exclusion criteria included previous chemotherapy by taxane for metastatic disease and previous anti-HER2 therapy. The study was done in accordance with Good Clinical Practice guidelines and the Declaration of Helsinki. All patients provided written informed consent. Approvals for the study protocol were obtained from the independent ethic committee (Clinical Ethics Committee, CHRU Besancon, 03/04/2019).

Treatment

Treatments were classified as TPFT (taxanes-platin-fluoropyrimidine-trastuzumab) regimen if patients received at least one cycle of this protocol, and standard (S) regimen if they received at least one cycle of platin-fluoropyrimidine-trastuzumab treatment with the exclusion of taxanes. TPFT regimen consisted of docetaxel (75 mg/m2 on day 1), cisplatin (75 mg/m2 on day 1), and 5-fluorouracil (750 mg/m2/day on continuous perfusion on days 1-5), every 3 weeks as previously described by Van Cutsem et al. (standard DCF regimen) or docetaxel (40 mg/m2 IV on day 1), cisplatin (40 mg/m2 IV on day 3) 5-fluorouracil (2000 mg/m2 intravenously over 48 hours) every 2 weeks as previously described by Shah et al. (modified DCF regimen).

Trastuzumab was given by intravenous infusion at a dose of 8mg/kg on day 1 of the first cycle, followed by 6 mg/kg every 3 weeks in association with standard DCF; or 6mg/kg on day 1 of the first cycle, followed by 4 mg/kg every 2 weeks in association with modified DCF. S regimen corresponds to the regimen defined in the ToGA trial or the combination of oxaliplatin and fluoropyrimidine (5-FU or capecitabine).

Conversion Therapy

In case of disease control rate (complete or partial response, or stable disease), a multidisciplinary team comprising medical oncologists and surgeons re-evaluated all patients to determine potential conversion therapy. This strategy was defined as a local treatment aiming at an R0 resection after chemotherapy in initially unresectable stage IV gastric cancer. In this study, it was composed of the resection of the primary tumor and local treatment (surgery, radiotherapy, cryotherapy, or radiofrequency) of metastasis.

Definition of Variables

The primary outcome was progression-free survival (PFS), defined as the time interval between the start of the treatment and the clinical or radiological progression, or death from any cause. Alive patients free of progression were censored at the last date of news. Clinical records were used to obtain baseline characteristics: gender, age at diagnosis, tumor localization, TNM classification (by the 2009 classification), characteristics of localized surgical management, (neo)adjuvant chemotherapy or radiotherapy, perioperative treatment, metastatic recurrence, and management.

Secondary outcomes were overall survival (OS), defined as the time interval between the start of the treatment and the death from any cause; the objective response rate (ORR) and the disease control rate (DCR) evaluated using Response Evaluation Criteria in Solid Tumors (RECIST) criteria version 1.1; the secondary resectability of primary tumor or metastasis in patients with DCR (complete or partial response, or stable disease); and the safety.

Statistical Analysis

Qualitative variables were described using frequency and percentage, and continuous variables were described using median and range. These characteristics were compared statistically using Chi2 or Fisher tests for categorical variables. Quantitative variables were compared statistically using a Wilcoxon test. Median follow-up with a 95% confidence interval (CI) was estimated with the reverse Kaplan Meier method. PFS and OS were assessed using a Kaplan Meier method, described with median and 95% confidence interval (CI), and the survivals between the two groups were compared using a log-rank test. Association between baseline characteristics and PFS and OS was investigated with Cox proportional hazards regression model and hazard ratio (HR) with 95%CI were provided. Variables with p-value < 0.15 in univariate analysis were entered in a multivariate model, and the treatment arm was forced. In order to limit bias due to potential confounding factors unbalanced between treatment groups, the inverse of probability of treatment weighting (IPTW) (24) method was applied in a univariate Cox model using the propensity score. This propensity score was derived from a multivariate logistic regression which estimates the probability to be in the T group using variables with p-value <0.15 in univariate logistic regression. Analysis of survival in the subgroup of patients’ ECOG PS 0-1 and synchronous metastasis was performed. We have also compared the secondary resectability between the two groups. Statistical analyses were performed under SAS version 9.4 considering a significance threshold α of 5%. These statistical analyses were carried out by the Methodology and Quality of Life in Cancerology Unit of the Besançon University Hospital (UMR 1098), based on data collected within the e-CRF.

Results

Patients’ Characteristics in the Overall Population

Clinical characteristics of the 65 evaluable patients are listed in Supplementary Table 1. Twenty-four patients (37%) received TPFT regimen (T group), and 41 patients (63%) received standard regimen (S group), with no taxanes. The median age was 68.7 (range, 30.3-90.6) and 75.4% had a synchronous metastatic disease at diagnosis. Sixty-three percent of patients had gastric tumors, and approximately 75% of the tumors have moderately or poorly differentiated histology. The population was relatively high functioning in both groups (ECOG performance status (PS) 0 or 1: 86% of patients). Nevertheless, 20% of patients presented PS of 2 on the ECOG scale in the S group, compared to 4.2% in the T group (p=0.14). Patients’ characteristics were similar in both groups, except for age [64.5 (39-79.3) in the T group versus 72.8 (30.3-90.6) for the S group (p=0.015)] and metastatic disease with no surgery of primary tumor (91.7% in T group vs 65.9% in S group, p=0.02) at diagnosis (Supplementary Table 1).

Selection of the Population of Analysis: Patients With ECOG PS 0-1 and Synchronous Metastasis

To study groups of patients with similar clinical characteristics, we decided to select patients with ECOG PS 0-1 and synchronous metastasis (Figure 2). Baseline characteristics of patients in the population of analysis were presented in Table 1; composed of 19 patients in the S group and 21 patients in the T group. The median age was 71.2 (range, 30.3-90.6) in the S group and 64.2 (range, 39-79.3) in the T group (p=0.035). Other patients’ characteristics were similar in both groups. In the case of taxanes based regimen, 16 patients received the mDCF regimen, 4 patients received the DCF regimen, and 1 patient received the FLOT regimen (Table 1).

FIGURE 2
www.frontiersin.org

Figure 2 Flowchart. S group: Standard regimen. T group: TPFT regimen. PS, Performance status.

TABLE 1
www.frontiersin.org

Table 1 Baseline characteristics of patients in the population of analysis.

Outcomes in the Population of Analysis

In patients with ECOG PS 0-1 and synchronous metastasis (the population of analysis), the median PFS was 9.3 months (95%CI 7.0 to 17.2) in the T group and 5.9 months (95%CI 3.7 to 9.6) in the S group (log-rank p=0.038) (Figure 3A). Results from univariate and multivariate Cox regression analysis are presented in Table 2. In this analysis, no clinical or pathological characteristics were associated with PFS. Treatment by taxanes was significantly associated with a better PFS in univariate (HR 0.49; 95%CI 0.25 to 0.98, p=0.042) and multivariate analysis (HR 0.44; 95%CI 0.21 to 0.94, p=0.033).

FIGURE 3
www.frontiersin.org

Figure 3 Outcomes according to the treatment group in the population of analysis. Population of analysis: patients with ECOG PS 0 - 1 and synchronous metastasis. (A) Progression-Free Survival and (B) Overall Survival. S group: Standard regimen. T group: TPFT regimen. CI, Confidence Interval.

TABLE 2
www.frontiersin.org

Table 2 Univariate and multivariate Cox regression for PFS in the population of analysis.

The median OS was 19.0 months (95%CI 7.8 to 45.2) in the T group and 13.0 months (95%CI 5.5 to 14.8) in the S group (log-rank p=0.033) (Figure 3B). In univariate Cox regression analysis, no baseline characteristics were associated with a better OS (Supplementary Table 2). Treatment by taxanes was significantly associated with a better OS (HR 0.45; 95%CI 0.21 to 0.96, p=0.0378) in univariate analysis, but not in multivariate analysis (HR 0.47; 95%CI 0.21 to 1.06, p=0.07).

Propensity Score

A propensity score, derived from multivariate logistic regression analysis was performed to estimate the probability to have taxanes-based chemotherapy in patients with synchronous metastasis and ECOG PS 0-1. Age and tumor localization were added in the multivariate logistic regression (Supplementary Table 3).

According to the IPTW method applied in univariate Cox analysis, the use of taxanes was associated with better PFS (HR 0.56; 95%CI 0.34 to 0.91, p=0.0187) and OS (HR 0,49; 95%CI 0.29 to 0.84, p=0.009) in the population of analysis (Table 3).

TABLE 3
www.frontiersin.org

Table 3 IPTW methodology applied in univariate Cox analysis for PFS and OS in the population of analysis.

Objective Response Rate and Conversion Therapy in the Population of Analysis

In patients with ECOG PS 0-1 and synchronous metastasis (the population of analysis), the objective response rate was 68.4% in the T group versus 47.4% in the S group (p=0.19) (Supplementary Table 4). In the T group, 5 patients had surgical resection of the primary tumor (29.4%) compared to 1 patient in the S group (p=0.35) (Supplementary Table 5). Median PFS was 26.8 months (95%CI 8.0 to not reached (NR)) in case of surgical resection of the primary tumor, and 8.5 months (95%CI 5.2 to 12.9) in patients without surgical resection (log-rank 0.032). Median OS was not reached (95%CI 17.4 to NR) in case of surgical resection of the primary tumor, versus 13.6 months (95%CI 7.2 to 22.5) in patients without surgical resection (log-rank 0.0992) (Figure 4). Characteristics of patients who underwent conversion surgery were described in Supplementary Table 6.

FIGURE 4
www.frontiersin.org

Figure 4 Outcomes according to conversion therapy in the T group of the population of analysis. Evaluation of progression-free survival and overall survival in the T group of the population of analysis (patients with ECOG PS 0 - 1 and synchronous metastasis). Conversion therapy corresponds to the resection of the primary tumor. Patients with conversion therapy have disease control after chemotherapy and local treatment of metastasis. CI, Confidence Interval; PFS, Progression-Free Survival; OS, Overall Survival.

Patients’ Exposure to Treatment and Toxicity in the Population of Analysis

In patients with ECOG PS 0-1 and synchronous metastasis, the median number of cycles was 8 (range, 2–16) in the T group compared to 9 in the S group (range, 1-15) (p=0.9566). The rate of patients with a dose reduction of one of the chemotherapy molecules during the entire course of treatment was 62% in the T group versus 74% in the S group (p=0.427). There was no difference in early dose reduction (cycle 1 to 3) between both groups (29% in the T group and 37% in the S group, p=0.577). No toxic death was observed in both groups (Supplementary Table 7).

Discussion

After the ToGA trial, the combination of platin-5-FU chemotherapy and trastuzumab became the standard of care in HER2-positive advanced GEA patients. In this study, we found that the combination of the addition of docetaxel to platin, 5-FU, and trastuzumab (TPFT) have a potential interest in selected patients. A survival advantage was observed in favor of the TPFT regimen, and this finding was consistent across different analytic approaches.

Despite the small number of patients, median PFS and OS in the S group remain comparable to the ToGA study, with 5.6 and 12.9 months, respectively in our study compared to 6.7 and 13.8 months in the ToGA study (1). In the S group, the ORR seems to be also similar, evaluated to 37% in our study and 47% in the ToGA study. In our T group, median PFS and OS were 9.3 and 19.0 months, respectively. Then, these results are comparable to single-arm small phase II trials evaluating taxanes, platin, 5-FU, and trastuzumab regimens (Table 4). The mPFS was 9.2 months to not reached, and mOS was 19.4 to not reached with a median follow up to 18.3 months (1921). More recently, a retrospective study including 22 patients treated with trastuzumab plus taxane-based chemotherapy, and 108 patients treated with trastuzumab plus standard chemotherapy as first-line treatment in HER2-positive advanced GEA patients, showed a significant improvement in OS in favor of taxanes. The mOS was 15.2 months (95% CI 12.7-17.7) versus 11.1 months (95% CI 8.3-13.9; p = 0.03) (25), in line with our results.

TABLE 4
www.frontiersin.org

Table 4 Main outcomes in patients with advanced HER-2 positive gastric cancer treated with taxanes-based chemotherapy and trastuzumab.

In our TanDHER study, an objective response was observed in 68.2% in the T group, similar to 60 to 94% of phase II trials (1921). Besides, the secondary resection of the primary tumor was performed in five among 17 patients (29%) in the T group, compared to one patient in the S group in the population of analysis. Mitsui et al. have also shown a high secondary resection rate with 9 of 16 patients (56%) undergoing surgery of their metastasis and primary tumor.

Over the last decade, published data are in favor of a conversion therapy after chemotherapy in selected patients and seems to offer longer survival than chemotherapy alone (26). For example, Yamaguchi et al. have shown that conversion therapy was performed in 77 patients in a retrospective study including in 259 advanced GEA patients with systemic chemotherapy. The median survival time was 30.5 months in patients who underwent surgical resection, as opposed to 11.3 months in those who received chemotherapy alone (27). In our study, we observed a prolonged PFS and OS in the case of surgical resection of the primary tumor. This therapeutic strategy seems to be an interesting option in advanced GEA and should be discussed in multidisciplinary board in good responders with oligometastatic disease.

Our study has several limits naturally related to its size and retrospectively analyzed results. However, our results are highly comparative with previous phase II and retrospective data and support the potential interest of the addition of taxane at first-line in metastatic GEA patients.

Besides, anti-PD-1 immunotherapy is a slowly gaining field in advanced GEA in first-line treatment in combination with chemotherapy. The recent results of the CheckMate 649 and ATTRACTION-4 clinical trials demonstrated efficacy in advanced HER2-negative GEA (28, 29). In HER2-positive GEA, phase II and III trials are still ongoing, evaluating the addition of anti-PD-1 therapy (as monotherapy or in combination with anti-CTLA-4) to trastuzumab +/- platin-based chemotherapy (NCT02901301, NCT03615326, NCT03409848) (30, 31). Recently, the prespecified intermediate analysis of Keynote 811 phase III study, demonstrated an improvement in ORR with the addition of pembrolizumab to the PFT regimen (74.4% vs 51.9%; p=0.00006) (30). Furthermore, the combination of immune checkpoints inhibitors to taxanes-based chemotherapy enhanced the anti-tumor activity in preclinical data. Recent findings describe their ability to stimulate innate immune response and to promote adaptive antitumor immune response, which can explain the additive effect of the two classes of drugs (32). Thus, the association of anti-PD-(L)1 monoclonal antibody with taxanes-based chemotherapy was approved for the first-line treatment of unresectable triple-negative breast cancer or metastatic squamous NSCLC (3335). In gastrointestinal oncology, a phase II study of DCFm regimen in combination with atezolizumab in unresectable locally advanced squamous cell anal carcinoma is still ongoing (36). At the interim analysis, no safety issue is described (publication under review). Based on these observations, adding anti-PD-(L)1 immunotherapy to trastuzumab plus taxanes-based chemotherapy could be considered for further investigations.

In conclusion, our data suggest that adding docetaxel to standard treatment in first-line advanced HER2 positive gastroesophageal adenocarcinoma is active in selective patients, with significantly longer PFS and OS compared to the standard regimen. Conversion therapy may be considered in oligometastatic patients with good responses.

Data Availability Statement

The original contributions presented in the study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding author.

Ethics Statement

The studies involving human participants were reviewed and approved by Clinical Ethics Committee, CHRU Besancon. The patients/participants provided their written informed consent to participate in this study.

Author Contributions

EO, JH, SK, and DV contributed to conception and design of the study. EO and SK organized the database. JH performed the statistical analysis. EO wrote the first draft of the manuscript. SK, JH, and DV wrote sections of the manuscript. All authors contributed to manuscript revision, read, and approved the submitted version.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Acknowledgments

We thank Dr. Marie Kroemer for the selection of patients. We thank Pr. Frederic Bibeau for his suggestions to reply to the comments of reviewers. We thank Sophie Depierre, Clinical Research Project Manager in the CHU of Besançon, for the help to write and configure the protocol of the TanDHER study.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2022.763926/full#supplementary-material

References

1. Bang Y-J, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki 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(9742):687–97. doi: 10.1016/S0140-6736(10)61121-X

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Makiyama A, Sukawa Y, Kashiwada T, Kawada J, Hosokawa A, Horie Y, et al. Randomized, Phase II Study of Trastuzumab Beyond Progression in Patients With HER2-Positive Advanced Gastric or Gastroesophageal Junction Cancer: WJOG7112G (T-ACT Study). JCO (2020) JCO.19.03077. doi: 10.1200/JCO.19.03077

CrossRef Full Text | Google Scholar

3. Satoh T, Xu R-H, Chung HC, Sun G-P, Doi T, Xu J-M, et al. Lapatinib Plus Paclitaxel Versus Paclitaxel Alone in the Second-Line Treatment of HER2 -Amplified Advanced Gastric Cancer in Asian Populations: TyTAN—A Randomized, Phase III Study. J Clin Oncol (2014) 32(19):2039–49. doi: 10.1200/JCO.2013.53.6136

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Thuss-Patience PC, Shah MA, Ohtsu A, Van Cutsem E, Ajani JA, Castro H, et al. Trastuzumab Emtansine Versus Taxane Use for Previously Treated HER2-Positive Locally Advanced or Metastatic Gastric or Gastro-Oesophageal Junction Adenocarcinoma (GATSBY): An International Randomised, Open-Label, Adaptive, Phase 2/3 Study. Lancet Oncol (2017) 18(5):640–53. doi: 10.1016/S1470-2045(17)30111-0

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Tabernero J, Hoff PM, Shen L, Ohtsu A, Shah MA, Cheng K, 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(10):1372–84. doi: 10.1016/S1470-2045(18)30481-9

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Shah MA, Xu R, Bang Y-J, Hoff PM, Liu T, Herráez-Baranda LA, et al. HELOISE: Phase IIIb Randomized Multicenter Study Comparing Standard-Of-Care and Higher-Dose Trastuzumab Regimens Combined With Chemotherapy as First-Line Therapy in Patients With Human Epidermal Growth Factor Receptor 2–Positive Metastatic Gastric or Gastroesophageal Junction Adenocarcinoma. JCO (2017) 35(22):2558–67. doi: 10.1200/JCO.2016.71.6852

CrossRef Full Text | Google Scholar

7. Shitara K, Bang Y-J, Iwasa S, Sugimoto N, Ryu M-H, Sakai D, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N Engl J Med (2020) 382(25):2419–30. doi: 10.1056/NEJMoa2004413

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Pectasides E, Stachler MD, Derks S, Liu Y, Maron S, Islam M, et al. Genomic Heterogeneity as a Barrier to Precision Medicine in Gastroesophageal Adenocarcinoma. Cancer Discov (2018) 8(1):37–48. doi: 10.1158/2159-8290.CD-17-0395

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Gambardella V, Fleitas T, Tarazona N, Cejalvo JM, Gimeno-Valiente F, Martinez-Ciarpaglini C, et al. Towards Precision Oncology for HER2 Blockade in Gastroesophageal Adenocarcinoma. Ann Oncol (2019) 30(8):1254–64. doi: 10.1093/annonc/mdz143

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Saeki H, Oki E, Kashiwada T, Arigami T, Makiyama A, Iwatsuki M, et al. Re-Evaluation of HER2 Status in Patients With HER2-Positive Advanced or Recurrent Gastric Cancer Refractory to Trastuzumab (KSCC1604). Eur J Cancer (2018) 105:41–9. doi: 10.1016/j.ejca.2018.09.024

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Van Cutsem E, Moiseyenko VM, Tjulandin S, Majlis A, Constenla M, Boni C, et al. Phase III Study of Docetaxel and Cisplatin Plus Fluorouracil Compared With Cisplatin and Fluorouracil As First-Line Therapy for Advanced Gastric Cancer: A Report of the V325 Study Group. J Clin Oncol (2006) 24(31):4991–7. doi: 10.1200/JCO.2006.06.8429

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Shah MA, Janjigian YY, Stoller R, Shibata S, Kemeny M, Krishnamurthi S, et al. Randomized Multicenter Phase II Study of Modified Docetaxel, Cisplatin, and Fluorouracil (DCF) Versus DCF Plus Growth Factor Support in Patients With Metastatic Gastric Adenocarcinoma: A Study of the US Gastric Cancer Consortium. J Clin Oncol (2015) 33(33):3874–9. doi: 10.1200/JCO.2015.60.7465

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Ajani JA, Gerdes H, Kleinberg LR, Park H. National Comprehensive Cancer Network. Gastric Cancer Version 4.2019 (2019). Available at: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1434 (Accessed December 20,2019).

Google Scholar

14. Pegram MD, Konecny GE, O’Callaghan C, Beryt M, Pietras R, Slamon DJ. Rational Combinations of Trastuzumab With Chemotherapeutic Drugs Used in the Treatment of Breast Cancer. JNCI J Natl Cancer Inst (2004) 96(10):739–49. doi: 10.1093/jnci/djh131

CrossRef Full Text | Google Scholar

15. Merlin J-L. In Vitro Comparative Evaluation of Trastuzumab (Herceptin(R)) Combined With Paclitaxel (Taxol(R)) or Docetaxel (Taxotere(R)) in HER2-Expressing Human Breast Cancer Cell Lines. Ann Oncol (2002) 13(11):1743–8. doi: 10.1093/annonc/mdf263

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Fujimoto-Ouchi K, Sekiguchi F, Yasuno H, Moriya Y, Mori K, Tanaka Y. Antitumor Activity of Trastuzumab in Combination With Chemotherapy in Human Gastric Cancer Xenograft Models. Cancer Chemother Pharmacol (2007) 59(6):795–805. doi: 10.1007/s00280-006-0337-z

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Di Modica M, Sfondrini L, Regondi V, Varchetta S, Oliviero B, Mariani G, et al. Taxanes Enhance Trastuzumab-Mediated ADCC on Tumor Cells Through NKG2D-Mediated NK Cell Recognition. Oncotarget (2016) 7(1):255–65. doi: 10.18632/oncotarget.6353

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Al-Batran S-E, Hozaeel W, Jäger E. Combination of Trastuzumab and Triple FLOT Chemotherapy (5-Fluorouracil/Leucovorin, Oxaliplatin, and Docetaxel) in Patients With HER2-Positive Metastatic Gastric Cancer: Report of 3 Cases. Onkologie (2012) 35(9):505–8. doi: 10.1159/000341841

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Mitsui Y, Sato Y, Miyamoto H, Fujino Y, Takaoka T, Miyoshi J, et al. Trastuzumab in Combination With Docetaxel/Cisplatin/S-1 (DCS) for Patients With HER2-Positive Metastatic Gastric Cancer: Feasibility and Preliminary Efficacy. Cancer Chemother Pharmacol (2015) 76(2):375–82. doi: 10.1007/s00280-015-2807-7

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Roviello G, Petrioli R, Nardone V, Rosellini P, Multari AG, Conca R, et al. Docetaxel, Oxaliplatin, 5FU, and Trastuzumab as First-Line Therapy in Patients With Human Epidermal Receptor 2- Positive Advanced Gastric or Gastroesophageal Junction Cancer: Preliminary Results of a Phase II Study. Medicine (2018) 97(20):e10745. doi: 10.1097/MD.0000000000010745

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Mondaca S, Margolis M, Sanchez-Vega F, Jonsson P, Riches JC, Ku GY, et al. Phase II Study of Trastuzumab With Modified Docetaxel, Cisplatin, and 5 Fluorouracil in Metastatic HER2-Positive Gastric Cancer. Gastric Cancer (2018) 22(2). doi: 10.1007/s10120-018-0861-7

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Hofmann M, Stoss O, Shi D, Büttner R, van de Vijver M, Kim W, et al. Assessment of a HER2 Scoring System for Gastric Cancer: Results From a Validation Study. Histopathology (2008) 52(7):797–805. doi: 10.1111/j.1365-2559.2008.03028.x

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Bartley AN, Washington MK, Colasacco C, Ventura CB, Ismaila N, Benson AB, et al. HER2 Testing and Clinical Decision Making in Gastroesophageal Adenocarcinoma: Guideline From the College of American Pathologists, American Society for Clinical Pathology, and the American Society of Clinical Oncology. JCO (2017) 35(4):446–64. doi: 10.1200/JCO.2016.69.4836

CrossRef Full Text | Google Scholar

24. Austin PC, Schuster T. The Performance of Different Propensity Score Methods for Estimating Absolute Effects of Treatments on Survival Outcomes: A Simulation Study. Stat Methods Med Res (2016) 25(5):2214–37. doi: 10.1177/0962280213519716

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Gürbüz M, Akkuş E, Sakin A, Urvay S, Demiray AG, Şahin S, et al. Combination of Trastuzumab and Taxane-Containing Intensified Chemotherapy in First-Line Treatment of HER2-Positive Advanced Gastric Cancer. Tumori (2020) 300891620969823.

Google Scholar

26. Zurleni T, Gjoni E, Altomare M, Rausei S. Conversion Surgery for Gastric Cancer Patients: A Review. World J Gastrointest Oncol (2018) 10(11):398–409. doi: 10.4251/wjgo.v10.i11.398

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Yamaguchi K, Yoshida K, Tanahashi T, Takahashi T, Matsuhashi N, Tanaka Y, et al. The Long-Term Survival of Stage IV Gastric Cancer Patients With Conversion Therapy. Gastric Cancer (2018) 21(2):315–23. doi: 10.1007/s10120-017-0738-1

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Boku N, Ryu M-H, Kato K, Chung HC, Minashi K, Lee K-W, et al. Safety and Efficacy of Nivolumab in Combination With S-1/Capecitabine Plus Oxaliplatin in Patients With Previously Untreated, Unresectable, Advanced, or Recurrent Gastric/Gastroesophageal Junction Cancer: Interim Results of a Randomized, Phase II Trial (ATTRACTION-4). Ann Oncol (2019) 30(2):250–8. doi: 10.1093/annonc/mdy540

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, 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(10294):27–40. doi: 10.1016/S0140-6736(21)00797-2

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Chung HC, Bang Y-J S, Fuchs C, Qin S-K, Satoh T, Shitara K, et al. First-Line Pembrolizumab/Placebo Plus Trastuzumab and Chemotherapy in HER2-Positive Advanced Gastric Cancer: KEYNOTE-811. Future Oncol (2021) 17(5):491–501. doi: 10.2217/fon-2020-0737

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Tintelnot J, Goekkurt E, Binder M, Thuss-Patience P, Lorenzen S, Knorrenschild JR, et al. Ipilimumab or FOLFOX With Nivolumab and Trastuzumab in Previously Untreated HER2-Positive Locally Advanced or Metastatic EsophagoGastric Adenocarcinoma - the Randomized Phase 2 INTEGA Trial (AIO STO 0217). BMC Cancer (2020) 20(1):503. doi: 10.1186/s12885-020-06958-3

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Serpico AF, Visconti R, Grieco D. Exploiting Immune-Dependent Effects of Microtubule-Targeting Agents to Improve Efficacy and Tolerability of Cancer Treatment. Cell Death Dis (2020) 11(5):361. doi: 10.1038/s41419-020-2567-0

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N Engl J Med (2018) 379(22):2108–21. doi: 10.1056/NEJMoa1809615

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N Engl J Med (2018) 378(24):2288–301. doi: 10.1056/NEJMoa1716948

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gümüş M, Mazières J, et al. Pembrolizumab Plus Chemotherapy for Squamous Non–Small-Cell Lung Cancer. N Engl J Med (2018) 379(21):2040–51. doi: 10.1056/NEJMoa1810865

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Kim S, Buecher B, André T, Jary M, Bidard F-C, Ghiringhelli F, et al. Atezolizumab Plus Modified Docetaxel-Cisplatin-5-Fluorouracil (mDCF) Regimen Versus mDCF in Patients With Metastatic or Unresectable Locally Advanced Recurrent Anal Squamous Cell Carcinoma: A Randomized, Non-Comparative Phase II SCARCE GERCOR Trial. BMC Cancer (2020) 20(1):352. doi: 10.1186/s12885-020-06841-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: taxanes, HER2, gastric carcinoma, metastatic, trastuzumab, survival

Citation: Orillard E, Henriques J, Vernerey D, Almotlak H, Calcagno F, Fein F, Fratté S, Jary M, Klajer E, Vienot A, Borg C and Kim S (2022) Interest of the Addition of Taxanes to Standard Treatment in First-Line Advanced HER2 Positive Gastroesophageal Adenocarcinoma in Selective Patients. Front. Oncol. 12:763926. doi: 10.3389/fonc.2022.763926

Received: 24 August 2021; Accepted: 03 January 2022;
Published: 07 March 2022.

Edited by:

Jia Wei, Nanjing Drum Tower Hospital, China

Reviewed by:

Ravindra Deshpande, Wake Forest School of Medicine, United States
Jane Rogers, University of Texas MD Anderson Cancer Center, United States
Anju Kumari, National Cancer Institute, United States

Copyright © 2022 Orillard, Henriques, Vernerey, Almotlak, Calcagno, Fein, Fratté, Jary, Klajer, Vienot, Borg and Kim. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Emeline Orillard, eorillard@chu-besancon.fr

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.