1
|
Svrcek M, Voron T, André T, Smyth EC, de la Fouchardière C. Improving individualised therapies in localised gastro-oesophageal adenocarcinoma. Lancet Oncol 2024; 25:e452-e463. [PMID: 39214116 DOI: 10.1016/s1470-2045(24)00180-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/05/2024] [Accepted: 03/20/2024] [Indexed: 09/04/2024]
Abstract
Despite our increased understanding of the biological and molecular aspects of gastro-oesophageal tumourigenesis, the identification of prognostic or predictive factors remains challenging. Patients with resectable gastric and oesophageal adenocarcinoma are often treated similarly after surgical resection, regardless of their tumour biology, clinical characteristics, and histological treatment response. Substantial progress has been made in the past 5 years in managing patients with gastric or oesophageal adenocarcinoma, including the use of immune checkpoint inhibitors and new targeted therapies, leading to substantial improvements in clinical outcomes. These advancements have primarily been established in advanced and metastatic disease, while the management framework for local and locoregional disease is just beginning to shift. We provide an overview of existing data on biomarkers and tumour-related and host-related factors that are relevant to stratify patients into low-risk and high-risk recurrence groups, both before and after surgery, paving the way for more personalised treatment approaches.
Collapse
Affiliation(s)
- Magali Svrcek
- Department of Pathology, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | - Thibault Voron
- Digestive Surgery Department, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | - Thierry André
- Department of Medical Oncology, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | | | | |
Collapse
|
2
|
Taniyama Y, Murakami K, Yoshida N, Takahashi K, Matsubara H, Baba H, Kamei T. Evaluating the effect of Neoadjuvant chemotherapy for esophageal Cancer using the RECIST system with shorter-axis measurements: a retrospective multicenter study. BMC Cancer 2021; 21:1008. [PMID: 34496769 PMCID: PMC8428108 DOI: 10.1186/s12885-021-08747-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/29/2021] [Indexed: 11/16/2022] Open
Abstract
Background Evaluating the effect on primary lesions is important in determining treatment strategies for esophageal cancer. The Response Evaluation Criteria in Solid Tumors system, which employs the longest diameter for measuring tumors, is commonly used for evaluating treatment effects. However, the usefulness of these criteria in assessing primary esophageal tumors remains controversial. Thus, we evaluated this issue by measuring not only the longest diameter but also the shorter axis of the tumor. Methods We retrospectively reviewed data from 313 patients with esophageal cancer treated with neoadjuvant chemotherapy followed by esophagectomy at three major high-volume centers in Japan. All patients underwent contrast-enhanced computed tomography before and after chemotherapy. The longest and shortest tumor diameters were measured in each case. Treatment effects were adapted to the Response Evaluation Criteria in Solid Tumors system. Correlations between pathological and survival data were also analyzed. Results Inter-observer discrepancies were examined for changes in the longest diameter and shorter axis of the tumor (the intraclass correlation coefficients were 0.550 and 0.624, respectively). The shorter axis was correlated with the pathological response in the multivariate analysis (p < 0.001). The shorter axis was significantly associated with overall survival and disease-free survival (both p < 0.001), whereas this association was not observed for the longest tumor diameter. Conclusions This multicenter study demonstrated that the Response Evaluation Criteria in Solid Tumors system is useful for predicting pathological response and survival by incorporating the shorter axis of the primary esophageal tumor. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08747-y.
Collapse
Affiliation(s)
- Yusuke Taniyama
- Department of Surgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Kentaro Murakami
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kozue Takahashi
- Department of Surgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.,Department of Radiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takashi Kamei
- Department of Surgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
3
|
Abstract
BACKGROUND At the time of diagnosis of gastric cancer approximately one third of patients already have metastases. It is important to differentiate between oligometastasis and the diffuse metastatic situation. For the first time the definition of oligometastasis has been integrated into the German S3 guidelines. OBJECTIVE Can multimodal treatment with tumor resection and metastasectomy combined with perioperative chemotherapy, increase the chances of survival in oligometastatic patients? MATERIAL AND METHODS In this review article the data situation of the current literature is discussed. RESULTS The Dutch D1/D2 trial reported an increased median survival for a subgroup of patients with single metastasis who underwent resection. Multimodal treatment with resection doubled the median survival of oligometastatic patients in the German AIO-FLOT 3 study and as a consequence, the AIO-FLOT 5 (RENAISSANCE) trial was designed. Patients with oligometastatic gastric and esophagogastric junction cancer are randomized after chemotherapy to either undergo resection followed by adjuvant chemotherapy or to undergo definitive chemotherapy. Further randomized trials investigate the benefit of antibodies and immune checkpoint inhibitors in locoregional and advanced metastatic gastric cancer with promising results. CONCLUSION The results of the ongoing randomized trials will show if oligometastatic patients benefit from a multimodal treatment with resection. The clear definition of the oligometastatic state, assessment of the response to neoadjuvant chemotherapy and realistic estimation of the R0 resectability will be useful for patient selection.
Collapse
|
4
|
Kadota T, Abe S, Yoda Y, Yoshinaga S, Oda I, Kojima T, Kato K, Daiko H, Yano T. Clinical outcomes according to the modified endoscopic criteria for neoadjuvant chemotherapy in resectable esophageal squamous cell carcinoma. Dig Endosc 2020; 32:337-345. [PMID: 31295769 DOI: 10.1111/den.13483] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/03/2019] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Neoadjuvant chemotherapy (nCT) followed by surgery is one of the standard treatments for resectable esophageal squamous cell carcinoma (ESCC). According to the Response Evaluation Criteria in Solid Tumors, endoscopic evaluation of a primary lesion is not recommended during nCT because of reduced objectivity. This study aimed to develop and validate endoscopic evaluation criteria for nCT. METHODS This study retrospectively investigated patients with T2/3 ESCC who underwent nCT followed by radical esophagectomy across two institutions (test and validation sets). We retrospectively estimated the therapeutic effect by classifying patients according to degree of tumor shrinkage (evaluated with endoscopy) as follows: marked reduction (MR), half reduction (HR), insufficient reduction (IR), and progressive disease (PD). Three endoscopists evaluated patients in the test set. Another three endoscopists evaluated patients in the validation set. We analyzed recurrence-free survival (RFS) 3 years after surgery. RESULTS Of 129 patients in the test set, 44 had MR, 35 had HR, 44 had IR, and six had PD. The 3-year RFS rates were 55% (overall), 79% (MR), 54% (HR), 35% (IR), and 33% (PD). Of 91 patients in the validation set, 22 had MR, 49 had HR, 18 had IR, and two had PD. The 3-year RFS rates were 54% (overall), 77% (MR), 55% (HR), 22% (IR), and 50% (PD). CONCLUSIONS Our endoscopic criteria for nCT predicted prognosis; however, future studies are needed to further investigate our criteria before general application in the clinical setting.
Collapse
Affiliation(s)
- Tomohiro Kadota
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Chiba, Japan
| | - Seiichiro Abe
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| | - Yusuke Yoda
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Chiba, Japan
| | | | - Ichiro Oda
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Kojima
- Department of Gastroenterology, Gastrointestinal Oncology Division, National Cancer Center Hospital East, Chiba, Japan
| | - Ken Kato
- Gastrointestinal Medical Oncology Divisions, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroyuki Daiko
- Department of Esophageal Surgery, National Cancer Center Hospital East, Chiba, Japan.,Esophageal Surgery Division, National Cancer Center Hospital, Tokyo, Japan
| | - Tomonori Yano
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Chiba, Japan
| |
Collapse
|
5
|
Yasufuku I, Nunobe S, Ida S, Kumagai K, Ohashi M, Hiki N, Sano T. Conversion therapy for peritoneal lavage cytology-positive type 4 and large type 3 gastric cancer patients selected as candidates for R0 resection by diagnostic staging laparoscopy. Gastric Cancer 2020; 23:319-327. [PMID: 31350702 DOI: 10.1007/s10120-019-00994-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The long-term outcomes of type 4 and large type 3 gastric cancer patients with positive peritoneal lavage cytology (CY1) remain unsatisfying. We evaluated our treatment strategy of conversion therapy for CY1 patients without peritoneal dissemination (P0). METHODS Diagnostic staging laparoscopy (DSL) was performed before treatment. Chemotherapy was applied for DSL-diagnosed P0CY1. The re-evaluation of peritoneal metastasis by staging laparoscopy (re-SL) was performed when a response to chemotherapy was identified by gastroscopy and/or CT. Gastrectomy with radical lymphadenectomy was applied as conversion therapy when peritoneal lavage cytology-negative (CY0) and P0 were diagnosed with re-SL, with the aim of achieving R0 resection. Chemotherapy was continued as palliative treatment in patients for whom re-SL was not applicable or when re-SL did not confirm P0CY0. The long-term outcomes were retrospectively evaluated. RESULTS Between 2009 and 2015, 214 patients with type 4 and large type 3 gastric cancer underwent DSL in the Cancer Institute Hospital. Thirty-nine patients were initially diagnosed with P0CY1. Seven patients received palliative gastrectomy first due to outlet obstruction or other reasons. Thirty-two patients received chemotherapy first. Among them, 13 patients underwent gastrectomy as conversion therapy and 19 were treated with palliative chemotherapy. The 3-year survival rate of patients who underwent conversion therapy, palliative chemotherapy and palliative gastrectomy was 76.9% [95% confidence interval (CI) 47.8-92.4%], 10.5% (95% CI 1.9-42.3%), and 0%, respectively. CONCLUSION Conversion therapy might be a promising treatment for P0CY1 type 4 and large type 3 gastric cancer patients. Re-SL was useful for selecting candidates for R0 resection.
Collapse
Affiliation(s)
- Itaru Yasufuku
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8551, Japan
| | - Souya Nunobe
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8551, Japan.
| | - Satoshi Ida
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8551, Japan
| | - Koshi Kumagai
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8551, Japan
| | - Manabu Ohashi
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8551, Japan
| | - Naoki Hiki
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8551, Japan
| | - Takeshi Sano
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8551, Japan
| |
Collapse
|
6
|
PET in Gastrointestinal, Pancreatic, and Liver Cancers. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
7
|
Neoadjuvant therapy for locally advanced gastric cancer patients. A population pharmacodynamic modeling. PLoS One 2019; 14:e0215970. [PMID: 31071108 PMCID: PMC6508715 DOI: 10.1371/journal.pone.0215970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/05/2019] [Indexed: 01/27/2023] Open
Abstract
Background Perioperative chemotherapy (CT) or neoadjuvant chemoradiotherapy (CRT) in patients with locally advanced gastric (GC) or gastroesophageal junction cancer (GEJC) has been shown to improve survival compared to an exclusive surgical approach. However, most patients retain a poor prognosis due to important relapse rates. Population pharmacokinetic-pharmacodynamic (PK/PD) modeling may allow identifying at risk-patients. We aimed to develop a mechanistic PK/PD model to characterize the relationship between the type of neoadjuvant therapy, histopathologic response and survival times in locally advanced GC and GEJC patients. Methods Patients with locally advanced GC and GEJC treated with neoadjuvant CT with or without preoperative CRT were analyzed. Clinical response was assessed by CT-scan and EUS. Pathologic response was defined as a reduction on pTNM stage compared to baseline cTNM. Metastasis development risk and overall survival (OS) were described using the population approach with NONMEM 7.3. Model evaluation was performed through predictive checks. Results A low correlation was observed between clinical and pathologic TNM stage for both T (R = 0.32) and N (R = 0.19) categories. A low correlation between clinical and pathologic response was noticed (R = -0.29). The OS model adequately described the observed survival rates. Disease recurrence, cTNM stage ≥3 and linitis plastica absence, were correlated to a higher risk of death. Conclusion Our model adequately described clinical response profiles, though pathologic response could not be predicted. Although the risk of disease recurrence and survival were linked, the identification of alternative approaches aimed to tailor therapeutic strategies to the individual patient risk warrants further research.
Collapse
|
8
|
Cosgrove ND, Mullady DK. Endoscopic evaluation of the esophageal cancer patient after chemoradiotherapy for persistent/recurrent cancer. Dis Esophagus 2018; 31:5040371. [PMID: 29931309 DOI: 10.1093/dote/doy023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Endoscopy has an important role in the pre- and post-treatment staging of esophageal cancer. Complete pathologic response following neoadjuvant chemoradiation therapy occurs in approximately 25% of patients. However, the ability to accurately detect this preoperatively with currently available endoscopic modalities is limited such that the default pathway is for fit patients to proceed with surgical resection. This article discusses the available endoscopic modalities (primarily Esophagogastroduodenoscopy [EGD] with mucosal biopsies and endoscopic ultrasonography with or without fine needle aspiration) used for post-treatment staging of esophageal cancer. We present data regarding the benefits and limitations of endoscopic methods in assessing for residual disease. Unfortunately, endoscopic modalities are not accurate enough to identify complete pathological responsers who may avoid surgical resection.
Collapse
Affiliation(s)
- N D Cosgrove
- Division of Gastroenterology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - D K Mullady
- Division of Gastroenterology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
9
|
Tumor regression grading of gastrointestinal cancers after neoadjuvant therapy. Virchows Arch 2017; 472:175-186. [PMID: 28918544 DOI: 10.1007/s00428-017-2232-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/28/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023]
Abstract
Neoadjuvant therapy has been successfully introduced in the treatment of locally advanced gastrointestinal malignancies, particularly esophageal, gastric, and rectal cancers. The effects of preoperative chemo- or radiochemotherapy can be determined by histopathological investigation of the resection specimen following this treatment. Frequent histological findings after neoadjuvant therapy include various amounts of residual tumor, inflammation, resorptive changes with infiltrates of foamy histiocytes, foreign body reactions, and scarry fibrosis. Several tumor regression grading (TRG) systems, which aim to categorize the amount of regressive changes after cytotoxic treatment in primary tumor sites, have been proposed for gastroesophageal and rectal carcinomas. These systems primarily refer to the amount of therapy-induced fibrosis in relation to the residual tumor (e.g., the Mandard, Dworak, or AJCC systems) or the estimated percentage of residual tumor in relation to the previous tumor site (e.g., the Becker, Rödel, or Rectal Cancer Regression Grading systems). TRGs provide valuable prognostic information, as in most cases, complete or subtotal tumor regression after neoadjuvant treatment is associated with better patient outcomes. This review describes the typical histopathological findings after neoadjuvant treatment, discusses the most commonly used TRG systems for gastroesophageal and rectal carcinomas, addresses the limitations and critical issues of tumor regression grading in these tumors, and describes the clinical impact of TRG.
Collapse
|
10
|
Goel R, Subramaniam RM, Wachsmann JW. PET/Computed Tomography Scanning and Precision Medicine: Esophageal Cancer. PET Clin 2017; 12:373-391. [PMID: 28867110 DOI: 10.1016/j.cpet.2017.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Esophageal cancer commonly has a poor prognosis, which requires an accurate diagnosis and early treatment to improve outcome. Other modalities for staging, such as endoscopic ultrasound imaging and computed tomography (CT) scans, have a role in diagnosis and staging. However, PET with fluorine-18 fluoro-2-deoxy-d-glucose/CT (FDG PET/CT) scanning allows for improved detection of distant metastatic disease and can help to prevent unnecessary interventions that would increase morbidity. FDG PET/CT scanning is valuable in the neoadjuvant chemotherapy assessment and predicting survival outcomes subsequent to surgery. FDG PET/CT scanning detects recurrent disease and metastases in follow-up.
Collapse
Affiliation(s)
- Reema Goel
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA
| | - Rathan M Subramaniam
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA; Department of Clinical Sciences, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA; Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA; Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA
| | - Jason W Wachsmann
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA.
| |
Collapse
|
11
|
Role of histological regression grade after two neoadjuvant approaches with or without radiotherapy in locally advanced gastric cancer. Br J Cancer 2016; 115:655-63. [PMID: 27537382 PMCID: PMC5023782 DOI: 10.1038/bjc.2016.252] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/24/2016] [Accepted: 07/19/2016] [Indexed: 12/17/2022] Open
Abstract
Background: The degree of histopathological response after neoadjuvant therapy in locally advanced gastric cancer (GC) is a key determinant of patients' long-term outcome. We aimed to assess the pattern of histopathological regression after two neoadjuvant approaches and its impact on survival times. Methods: Regression grade of the primary tumour (Becker criteria) and the degree of nodal response by a 4-point scale (grades A–D) were assessed. Grade A—true negative lymph nodes (LNs); grade B and C—infiltrated LNs with any or little evidence of nodal response; and grade D—complete pathological response in a previously infiltrated LN. A favourable pathological response was defined as Becker Ia–b and grade D. Results: From 2004 to 2014, 80 patients with GC (cT3–4/N+ by CT-scan/EUS) were treated with either preoperative chemotherapy (ChT, n=34) or chemoradiation (CRT, n=46). Patients in the CRT group had a higher likelihood of achieving a Becker Ia–b response (58 vs 32%, P=0.001), a grade D nodal regression (30 vs 6%, P=0.009) and a favourable pathological response (23 vs 3% P=0.019). Patients with a grade D nodal response had a longer 5-year PFS and OS compared with those with a grade B or C response. Patients with a baseline negative LN status had similar outcomes irrespective of the preoperative therapy received (5-year OS; ChT vs CRT, 58 vs 51%, P=0.92). Conclusions: Preoperative chemoradiation increases the likelihood of achieving favourable histopathological features that correlate with a 5-year OS>70% in GC patients.
Collapse
|
12
|
Koerfer J, Kallendrusch S, Merz F, Wittekind C, Kubick C, Kassahun WT, Schumacher G, Moebius C, Gaßler N, Schopow N, Geister D, Wiechmann V, Weimann A, Eckmann C, Aigner A, Bechmann I, Lordick F. Organotypic slice cultures of human gastric and esophagogastric junction cancer. Cancer Med 2016; 5:1444-1453. [PMID: 27073068 PMCID: PMC4944870 DOI: 10.1002/cam4.720] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 02/21/2016] [Accepted: 03/08/2016] [Indexed: 12/19/2022] Open
Abstract
Gastric and esophagogastric junction cancers are heterogeneous and aggressive tumors with an unpredictable response to cytotoxic treatment. New methods allowing for the analysis of drug resistance are needed. Here, we describe a novel technique by which human tumor specimens can be cultured ex vivo, preserving parts of the natural cancer microenvironment. Using a tissue chopper, fresh surgical tissue samples were cut in 400 μm slices and cultivated in 6-well plates for up to 6 days. The slices were processed for routine histopathology and immunohistochemistry. Cytokeratin stains (CK8, AE1/3) were applied for determining tumor cellularity, Ki-67 for proliferation, and cleaved caspase-3 staining for apoptosis. The slices were analyzed under naive conditions and following 2-4 days in vitro exposure to 5-FU and cisplatin. The slice culture technology allowed for a good preservation of tissue morphology and tumor cell integrity during the culture period. After chemotherapy exposure, a loss of tumor cellularity and an increase in apoptosis were observed. Drug sensitivity of the tumors could be assessed. Organotypic slice cultures of gastric and esophagogastric junction cancers were successfully established. Cytotoxic drug effects could be monitored. They may be used to examine mechanisms of drug resistance in human tissue and may provide a unique and powerful ex vivo platform for the prediction of treatment response.
Collapse
Affiliation(s)
- Justus Koerfer
- Institute for AnatomyUniversity Medicine LeipzigLiebigstraße 1304103LeipzigGermany
- University Cancer Center Leipzig (UCCL)University Medicine LeipzigLiebigstraße 2004103LeipzigGermany
| | - Sonja Kallendrusch
- Institute for AnatomyUniversity Medicine LeipzigLiebigstraße 1304103LeipzigGermany
| | - Felicitas Merz
- Institute for AnatomyUniversity Medicine LeipzigLiebigstraße 1304103LeipzigGermany
| | - Christian Wittekind
- Institute of PathologyUniversity Medicine LeipzigLiebigstraße 2404103LeipzigGermany
| | - Christoph Kubick
- Institute of PathologyUniversity Medicine LeipzigLiebigstraße 2404103LeipzigGermany
| | - Woubet T. Kassahun
- Department for Visceral, Transplantation Thoracic and Vascular SurgeryUniversity Medicine LeipzigLiebigstraße 2004103LeipzigGermany
| | - Guido Schumacher
- Department for General and Visceral SurgeryKlinikum BraunschweigSalzdahlumer Straße 9038126BraunschweigGermany
| | - Christian Moebius
- Department for General and Visceral SurgeryKlinikum BraunschweigSalzdahlumer Straße 9038126BraunschweigGermany
| | - Nikolaus Gaßler
- Institute of PathologyKlinikum BraunschweigCeller Straße 3838114BraunschweigGermany
| | - Nikolas Schopow
- Institute for AnatomyUniversity Medicine LeipzigLiebigstraße 1304103LeipzigGermany
| | - Daniela Geister
- Institute of PathologyKlinikum St. GeorgDelitzscher Str. 14104129LeipzigGermany
| | - Volker Wiechmann
- Institute of PathologyKlinikum St. GeorgDelitzscher Str. 14104129LeipzigGermany
| | - Arved Weimann
- Department for General and Visceral SurgeryKlinikum St. GeorgDelitzscher Str. 14104129LeipzigGermany
| | - Christian Eckmann
- Department for General, Visceral and Thoracic SurgeryKlinikum PeineVirchowstraße 831226PeineGermany
| | - Achim Aigner
- Rudolf‐Boehm‐Institute for Pharmacology and ToxicologyClinical PharmacologyUniversity Medicine LeipzigHärtelstraße 16‐1804107LeipzigGermany
| | - Ingo Bechmann
- Institute for AnatomyUniversity Medicine LeipzigLiebigstraße 1304103LeipzigGermany
| | - Florian Lordick
- University Cancer Center Leipzig (UCCL)University Medicine LeipzigLiebigstraße 2004103LeipzigGermany
| |
Collapse
|
13
|
Blank S, Knebel P, Haag GM, Bruckner T, Klaiber U, Burian M, Schaible A, Sisic L, Schmidt T, Diener MK, Ott K. Immediate tumor resection in patients with locally advanced gastroesophageal adenocarcinoma with nonresponse to chemotherapy after 4 weeks of treatment versus resection after completion of chemotherapy (OPTITREAT trial, DRKS00004668): study protocol for a randomized controlled pilot trial. Pilot Feasibility Stud 2016; 2:18. [PMID: 27965838 PMCID: PMC5153833 DOI: 10.1186/s40814-016-0059-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 03/08/2016] [Indexed: 12/22/2022] Open
Abstract
Background Neoadjuvant chemotherapy is a standard of care for patients with adenocarcinoma of the esophagus and stomach in Europe, but still only 20–40 % respond to therapy and the critical issue; how to treat nonresponding patients is still unclear. So far, there is no randomized trial evaluating the impact of early termination of neoadjuvant chemotherapy and immediate tumor resection in nonresponding patients with locally advanced gastroesophageal cancer on postoperative outcome. With this exploratory pilot trial, we want to get first estimates about the effect of discontinuation of chemotherapy with the aim to plan and conduct a further definitive trial. Methods/design OPTITREAT is designed as a single-center, randomized controlled pilot trial with two parallel study groups. Four weeks after starting neoadjuvant chemotherapy in all patients, clinical response will be assessed by endoscopy and endosonographic ultrasound. Then, nonresponding patients (n = 84) will be randomized in a 1:1 ratio to intervention group with stopping chemotherapy and immediate tumor resection or control group with completion of chemotherapy before surgery. Outcome measures are overall survival, R0 resection rate, perioperative morbidity and mortality, histopathological response, and quality of life. Statistical analysis will be based on the intention-to-treat population. Due to the study design as an explorative pilot trial, no formal sample size calculation was performed. The planned total sample size of 120 patients is considered ethical and large enough to show the feasibility and safety of the concept. First data on differences between the study groups in the defined endpoints will also be generated. Discussion Individualized therapy is of utmost interest in the treatment of locally advanced gastroesophageal adenocarcinoma as less than half of the patients show objective response to current chemotherapy regimens. The findings of the OPTITREAT trial will help to get first data about clinical response evaluation followed by immediate tumor resection in nonresponding patients after 4 weeks of neoadjuvant chemotherapy. Based on the results of this pilot study, a future confirmatory trial will be planned to prove efficacy and evaluate significance. Trial registration German Clinical Trial Register number: DRKS00004668
Collapse
Affiliation(s)
- Susanne Blank
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, Heidelberg, 69120 Germany
| | - Phillip Knebel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, Heidelberg, 69120 Germany.,Study Centre of the German Surgical Society (SDGC), University of Heidelberg, Im Neuenheimer Feld 110, Heidelberg, 69120 Germany
| | - Georg-Martin Haag
- Department of Medical Oncology, National Center of Tumor Diseases, University Hospital Heidelberg, Im Neuenheimer Feld 460, Heidelberg, 69120 Germany
| | - Thomas Bruckner
- Institute of Medical Statistics and Informatics, University of Heidelberg, Im Neuenheimer Feld 305, Heidelberg, 69120 Germany
| | - Ulla Klaiber
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, Heidelberg, 69120 Germany
| | - Maria Burian
- Department of General Visceral and Transplantation Surgery, Endoscopic Center, University of Gießen, Rudolf-Buchheimstr. 7, Gießen, 35392 Germany
| | - Anja Schaible
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, Heidelberg, 69120 Germany.,Interdisciplinary Endoscopic Center, University of Heidelberg, Im Neuenheimer Feld 460, Heidelberg, 69120 Germany
| | - Leila Sisic
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, Heidelberg, 69120 Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, Heidelberg, 69120 Germany
| | - Markus K Diener
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, Heidelberg, 69120 Germany.,Study Centre of the German Surgical Society (SDGC), University of Heidelberg, Im Neuenheimer Feld 110, Heidelberg, 69120 Germany
| | - Katja Ott
- Department of Surgery, RoMed Klinikum, Pettenkoferstr. 10, Rosenheim, 83022 Germany
| |
Collapse
|
14
|
Aichler M, Motschmann M, Jütting U, Luber B, Becker K, Ott K, Lordick F, Langer R, Feith M, Siewert JR, Walch A. Epidermal growth factor receptor (EGFR) is an independent adverse prognostic factor in esophageal adenocarcinoma patients treated with cisplatin-based neoadjuvant chemotherapy. Oncotarget 2015; 5:6620-32. [PMID: 25216514 PMCID: PMC4196151 DOI: 10.18632/oncotarget.2268] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neoadjuvant platin-based therapy is accepted as a standard therapy for advanced esophageal adenocarcinoma (EAC). Patients who respond have a better survival prognosis, but still a significant number of responder patients die from tumor recurrence. Molecular markers for prognosis in neoadjuvantly treated EAC patients have not been identified yet. We investigated the epidermal growth factor receptor (EGFR) in prognosis and chemotherapy resistance in these patients. Two EAC patient cohorts, either treated by neoadjuvant cisplatin-based chemotherapy followed by surgery (n=86) or by surgical resection (n=46) were analyzed for EGFR protein expression and gene copy number. Data were correlated with clinical and histopathological response, disease-free and overall survival. In case of EGFR overexpression, the prognosis for neoadjuvant chemotherapy responders was poor as in non-responders. Responders had a significantly better disease-free survival than non-responders only if EGFR expression level (p=0.0152) or copy number (p=0.0050) was low. Comparing neoadjuvantly treated patients and primary resection patients, tumors of non-responder patients more frequently exhibited EGFR overexpression, providing evidence that EGFR is a factor for indicating chemotherapy resistance. EGFR overexpression and gene copy number are independent adverse prognostic factors for neoadjuvant chemotherapy-treated EAC patients, particularly for responders. Furthermore, EGFR overexpression is involved in resistance to cisplatin-based neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Michaela Aichler
- Research Unit Analytical Pathology- Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstraße 1, Neuherberg, Germany
| | - Martin Motschmann
- Research Unit Analytical Pathology- Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstraße 1, Neuherberg, Germany
| | - Uta Jütting
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstraße 1, Neuherberg, Germany
| | - Birgit Luber
- Institute of Pathology, Technische Universität München, Trogerstraße 18, München, Germany
| | - Karen Becker
- Institute of Pathology, Technische Universität München, Trogerstraße 18, München, Germany
| | - Katja Ott
- Department of Surgery, University Hospital of Heidelberg, Im Neuenheimer Feld 110, Heidelberg, Germany
| | - Florian Lordick
- University Cancer Center Leipzig, University Clinic Leipzig, Liebigstraße 20, Leipzig, Germany
| | - Rupert Langer
- Institute of Pathology, Technische Universität München, Trogerstraße 18, München, Germany
| | - Marcus Feith
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Straße 22, München, Germany
| | - Jörg Rüdiger Siewert
- Directorate, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Axel Walch
- Research Unit Analytical Pathology- Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstraße 1, Neuherberg, Germany
| |
Collapse
|