Skip to main content
Erschienen in: Annals of Surgical Oncology 1/2024

Open Access 30.09.2023 | Gastrointestinal Oncology

Potential Predictive Immune and Metabolic Biomarkers of Tumor Microenvironment Regarding Pathological and Clinical Response in Esophageal Cancer After Neoadjuvant Chemoradiotherapy: A Systematic Review

verfasst von: H. H. Wang, BSc, E. N. Steffens, BSc, G. Kats-Ugurlu, MD, PhD, B. van Etten, MD, PhD, J. G. M. Burgerhof, MSc, G. A. P. Hospers, MD, PhD, J. T. M. Plukker, MD, PhD

Erschienen in: Annals of Surgical Oncology | Ausgabe 1/2024

Abstract

Introduction

The tumor microenvironment (TME) plays a crucial role in therapy response and modulation of immunologic surveillance. Adjuvant immunotherapy has recently been introduced in post-surgery treatment of locally advanced esophageal cancer (EC) with residual pathological disease after neoadjuvant chemoradiotherapy (nCRT). F-18 fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG-PET/CT) remains a valuable imaging tool to assess therapy response and to visualize metabolic TME; however, there is still a paucity in understanding the interaction between the TME and nCRT response. This systematic review investigated the potential of TME biomarkers and 18F-FDG-PET/CT features to predict pathological and clinical response (CR) after nCRT in EC.

Methods

A literature search of the Medline and Embase electronic databases identified 4190 studies. Studies regarding immune and metabolic TME biomarkers and 18F-FDG-PET/CT features were included for predicting pathological response (PR) and/or CR after nCRT. Separate analyses were performed for 18F-FDG-PET/CT markers and these TME biomarkers.

Results

The final analysis included 21 studies—10 about immune and metabolic markers alone and 11 with additional 18F-FDG-PET/CT features. High CD8 infiltration before and after nCRT, and CD3 and CD4 infiltration after nCRT, generally correlated with better PR. A high expression of tumoral or stromal programmed death-ligand 1 (PD-L1) after nCRT was generally associated with poor PR. Moreover, total lesion glycolysis (TLG) and metabolic tumor volume (MTV) of the primary tumor were potentially predictive for clinical and PR.

Conclusion

CD8, CD4, CD3, and PD-L1 are promising immune markers in predicting PR, whereas TLG and MTV are potential 18F-FDG-PET/CT features to predict clinical and PR after nCRT in EC.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1245/​s10434-023-14352-z.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Standard curative treatment for potentially resectable locally advanced esophageal cancer (EC) (clinical TNM classification cT2-4a/Nany/M0) consists of neoadjuvant chemoradiotherapy (nCRT) followed by surgery according to the CROSS regimen.1 Although 20–30% of patients achieved a pathologic complete response (pCR), more than half still have residual tumor after nCRT.2 The tumor microenvironment (TME) is currently a focus in exploring additional treatment combinations. Several potential predictive markers have been evaluated to improve treatment outcome, including the expression of human epidermal growth factor receptor 2 (HER2) and programmed death-ligand 1 (PD-L1), as well as the prevalence of microsatellite instability (MSI). Based on the presence of MSI and PD-L1 expression among squamous cell (ESCC) and adenocarcinoma (EAC) of the esophagus, including the results of the landmark CheckMate-577 trial, adjuvant immunotherapy has been suggested in EC patients with residual pathologic disease.310 Differences in response of only immune checkpoint inhibitors have been observed in ESCC and EAC, which might be associated with important differences in the TME in upper/mid-esophageal (ESCC) and lower esophagus/gastroesophageal adenocarcinoma (EAC/GEA).1113 Compared with EAC/GEA, ESCC exhibited a high expression of PD-L1 and a low HER-2 expression and high MSI (MSI-H) status.14
Therapy response and the activities of the TME are commonly visualized with F-18 fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG-PET/CT) scanning. 18F-FDG-uptake (glucose analog) measured by PET/CT indicates the highly increased glucose uptake because of the Warburg effect in tumor tissue. Many studies showed that the increased uptake of glucose and glycolysis by esophageal tumor cells might be caused by enhancement of membrane-bound glucose transporters (GLUT) and hexokinase (HK) enzymes.1517 Studies have ever since tried to associate the Warburg effect in the tumor and its increased TME metabolic biomarkers with the semiquantitative standardized maximum uptake value (SUVmax) in 18F-FDG-PET/CT.18
However, there is still a gap in our understanding of how the TME interacts with nCRT in EC. Therefore, we performed a systematic review to explore potential metabolic and immune TME biomarkers and their predictive role in pathological response (PR) and/or clinical response (CR) after nCRT in EC. As 18F-FDG-PET/CT may visualize the metabolic activity throughout the entire tumor, including its inflammatory microenvironment, it can be used to study the effect of additional immunotherapy in future studies. Combined with potent biomarkers, this metabolic imaging may be helpful in determining response to identify patients more likely to benefit from additional treatment or a potentially applicable organ-preserving treatment approach. Therefore, we also aimed to provide some future research perspectives on metabolic and immune TME biomarkers that might be associated with 18F-FDG-PET/CT (semi)-quantitative features.

Materials and Methods

Search Strategy and Study Selection Process

A systematic review according to the Preferred Reporting Items for Systematic Review and Meta-analysis Protocols (PRISMA-P) guidelines was performed.19 The study protocol was registered and the search strategy was documented online at the International Prospective Register of Systematic Reviews Registry (PROSPERO; ID CRD42022325532). The research question was to explore potential predictive immune and metabolic biomarkers in the interaction of nCRT and TME for a more effective treatment strategy. The exact search strategy is provided in electronic supplementary material (ESM) Table 1. The EMBASE and PubMed online databases were searched from 2001 until September 2022 using the following inclusion criteria: (1) original article/conference abstracts; (2) studies on ESCC or EAC and/or GEA; (3) published in peer-reviewed journals from 2001 or later; (4) studies on the effect of the metabolic, immune and PET-based TME on PR and/or CR after neoadjuvant treatment; and (5) studies published in English. The exclusion criteria were (1) studies with missing or unclear description/criteria for groups and/or variables; (2) if full text was not available; (3) studies not assessing CR after nCRT on pre- and post-treatment PET/CT; and (4) studies not including pathologic reports of the esophageal biopsy and PR of the surgical resection material.
Table 1
Main characteristics of selected studies
ID
Author
Journal
Year
Study design
N
TNM staging
Histology
CT/CRT/RT
Tissue type
Pathological score
Clinical score
Time of scan
Metabolic/immune markers
1
Göbel et al.28 a
Clinical and Translational Radiation Oncology
2022
Retrospective
76
II–III
EAC, GEAC
CRT
Pretreatment biopsies
Mandard (TRG1–3 vs. TRG 4–5)
NA
NA
CD8, PD-1, PD-L1
2
Haddad et al.29 a
Journal of Personalized Medicine
2022
Retrospective
43
I–III
EAC, GEAC, ESCC
CRT;CT
Surgical specimens
College of American Pathologist (1–3)
NA
NA
CD3+, CD4+, CD8+, CD45R0+, Foxp3+, CD20+, CD163+
3
Fassan et al. 23 a
Cancer Medicine
2019
Retrospective
88
I–IV
ESCC
CRT
Surgical specimens
Mandard (TRG1 vs. TRG2–5)
NA
NA
CD4; Tbet; FoxP3; CD8; CD80; PD-L1;
4
Goedegebuure et al.24 a
Oncoimmunology
2021
Retrospective
40
II–III
EAC, GEAC
CRT
Tumor samples; surgical specimens
Mandard (TRG1 vs. TRG 2–3 vs. TRG4–5)
NA
NA
CD8/CD163; CK: CD3; FoxP3; CD163; Ki67
5
Huang et al.25 a
Journal of Clinical Medicine
2019
Retrospective
107
II–III
ESCC
CRT
Surgical specimens
pCR; no pCR
NA
NA
PD-L1
6
Koemans et al.30 a
Histopathology
2021
Retrospective
123
II–III
EAC, GEAC
CRT
Surgical specimens
Mandard (TRG2 vs. TRG4–5)
NA
NA
CD3+, CD4+, CD8+, FOXP3, PD-L1
7
Kotsafti et al.26 a
Oncoimmunology
2020
Prospective
123
II–III
EAC, GEAC
CRT
Pretreatment biopsies; healthy esophageal mucosa close to cancer site
Mandard (TRG1 vs. TRG2–5)
NA
NA
mRNA of CD80; CD8; CD28; CD38; CTLA4; CD8alpha; CD8beta; CD107a (LAMP1); CD69; Tbet (TBX21); SERPINB3; TP53; HER2 (ERBB2); PD-1 (PDCD1); PD-L1 (CD274); PD-L2 (PDCD1LG2); MLH1; MSH6; MSH3; PMS2; BRAF; IFNγ; FOXP3; CD25 (IL2RA); CD94 (KLRD1); CTLA4; TNFβ (LT; TNFSF1)
8
Soeratram et al.27 a
The Journal of Pathology
2021
Retrospective
188
II–III
EAC, GEAC
CRT
Pretreatment biopsies
Mandard (TRG1–3 vs. 4–5)
NA
NA
PD-L1; PD-1; CD274PD-1; CD279; FOXP3; CD8; pan-cytokeratin multiplex; MHC class I, II duplex
9
Alvarado et al.31 a
Seminars in Thoracic and Cardiovascular Surgery
2022
Retrospective
244
II–III
EAC, GEAC, ESCC
CRT
NA
pCR; no pCR
NA
NA
Diabetes
10
Boyd et al.41 a
ASCO Annual Meeting I
2010
Retrospective
122
NA
EAC, GEAC, ESCC
CRT
NA
pCR; no pCR
NA
NA
DM; obesity
11
Fang et al.32 b
International Journal of Radiation Oncology Biology Physics
2017
Prospective
20
IIa–IIIb
EAC; GEAC, ESCC
CRT
NA
Mandard (TRG1 vs. TRG2 vs. TRG3+)
ΔSUVmax; ΔSUVmean; ΔTLG; MTV
Pre-CRT; during CRT; post-CRT
NA
12
Gillham et al.33 b
British Journal of Cancer
2006
Retrospective
32
II–III
EAC, GEAC, ESCC
CRT
NA
Mandard (TRG1–2 vs. TRG3–5)
ΔSUVmean; MTV
Pre-CRT; during CRT
NA
13
Kukar et al.34 b
JAMA Surgery
2015
Retrospective
77
NA
EAC, GEAC
CRT
NA
pCR; no pCR
SUVmax; ΔSUVmean; %reduction SUVmax; mean linear length of uptake, in cm, pre- and post-CRT; change in linear length
Pre-CRT; post-CRT
NA
14
Arnett et al.35 b
International Journal of Radiation Oncology
2016
Retrospective
193
I–IV
EAC, GEAC
CRT
NA
pCR; no pCR
ΔSUVmax; ΔSUR to blood pool uptake; ΔSUR to liver uptake
Pre-CRT; post-CRT
NA
15
Choi et al.36 b
European Journal of Nuclear Medicine and Molecular Imaging
2021
Retrospective
480
0–III
ESCC
CRT
NA
Mandard (TRG1 vs. TRG2–5)
MTV; SUVmax; SUVavg; TLG
Pre-CRT; post-CRT
NA
16
Dewan et al.37 b
Journal of Gastrointestinal Cancer
2017
Prospective
70
I–III
ESCC
CRT
NA
pCR; no pCR
SUVmax; % ΔSUVmax
Pre-CRT; post-CRT
NA
17
Lee et al.38 b
European Journal of Cardio-Thoracic Surgery
2021
Retrospective
158
I–III
ESCC
CRT
NA
pCR; no pCR
ΔSUVmax; % ΔSUVmax
Pre-CRT; post-CRT
NA
18
Piessen et al.39 b
Annals of Surgery
2013
Retrospective
60
II–III
EAC, GEAC, ESCC
CRT
NA
Grade I: ≥50% VRTC; Grade II: 10–50% VRTC with fibrosis; Grade III: <10%42
% ΔSUVmax
Pre-CRT; post-CRT
NA
19
Van Rossum et al.40 b
European Journal of Nuclear Medicine and Molecular Imaging
2017
Retrospective
70
 
EAC, GEAC
CRT
NA
Chirieac’s TRG: TRG1–4 (TRG1–2 vs. TRG3–4)43
SUVmax; SUVmean; MTV; TLG;
Pre-CRT; post-CRT
NA
20
Li et al.21 b
Scientific Reports
2021
Retrospective
127
II–III
ESCC
CRT
Pre-CRT blood; post-CRT blood
pCR; no pCR
ΔSUV; ΔSUV ratio
Pre-CRT; post-CRT
NLR pre-CRT; NLR post-CRT
21
Wang et al.22 b
European Journal of Cancer
2010
Retrospective
405
I–III
EAC, GEAC, ESCC, other
CRT
Surgical specimens
pCR; no pCR
NA
Pre-CRT; post-CRT
Obese (≥25 kg/m2);
non-obese (< 25 kg/m2)
a No presence of 18F-FDG-PET scan
b Presence of 18F-FDG-PET scan
CT chemotherapy, CRT chemoradiotherapy, RT radiotherapy, EAC esophageal adenocarcinoma, GEAC gastroesophageal adenocarcinoma, ESCC esophageal squamous cell carcinoma, pCR pathologic complete response, SUVmax maximum standardized uptake value, SUVmean mean standardized uptake value, SUVavg average standardized uptake value, TLG total lesion glycolysis, MTV metabolic tumor volume, SUR standardized uptake ratio, TRG tumor regression grade, NLR neutrophil to lymphocyte ratio, NA not available, PD-L1 programmed death-ligand 1, PD-1 programmed death-1, MHC major histocompatibility complex, DM diabetes mellitus, 18F-FDG-PET/CT F-18 fluorodeoxyglucose positron emission tomography/computed tomography

Quality Assessment

Risk of bias was assessed according to the study design and purpose. Non-randomized intervention studies were assessed using the Cochrane Risk of Bias in Nonrandomized Studies of Interventions (ROBINS-I) tool.20 All studies were evaluated with a visualization tool for risk-of-bias assessments in a systematic review (Risk-of-Bias VISualization Tool). Each article was read and assessed by two independent authors (HHW, ENS).

Data Extraction and Synthesis

Two authors (HHW, ENS) extracted the data independently. Disagreements between individual judgments were resolved by discussion among the research group consisting of two surgical oncologists, one medical oncologist and one pathologist (all experienced) until consensus was reached. Data were recorded, extracted and managed in a Microsoft Excel spreadsheet (Microsoft Corporation, Redmond, WA, USA). The extraction and generation of the results were discussed together with a statistician (JGMB).
Relative and percentage ΔSUV, total lesion glycolysis (TLG) and metabolic tumor volume (MTV) changes were considered to be an index for CR on 18F-FDG-PET/CT scans.

Results

Identification of Studies

The initial electronic search identified 4190 studies. After eliminating duplicates, 3097 studies remained. These studies were screened using title and/or abstract to assess relevancy to our study scope. As both PR and CR were assessed, we distinguished between studies that included 18F-FDG-PET/CT scans and studies that did not. Seventy-eight articles were included for full screening (31 congress abstracts, 47 original articles); 57 were excluded due to unclear description/criteria for groups and/or variables (n = 34) or studies that did not assess PR and/or CR (n = 23). Finally, we included 21 studies (20 original articles2140 and one study congress abstract41). We identified 10 studies on biological immune and metabolic TME biomarkers without the presence of an 18F-FDG-PET/CT scan (two studies on metabolic biomarkers, eight on immune biomarkers). Eleven studies were considered significant on clinical immune and metabolic TME biomarkers with the presence of an 18F-FDG-PET/CT scan (10 studies on metabolic biomarkers and 1 study on immune biomarkers) (Fig. 1).

Study Characteristics

An overview of the study characteristics of the selected studies is provided in Table 1. Three studies26,32,37 were prospective and 18 studies were retrospective.2124,2631,3336, 3841 Eight studies assessed only EAC,24,2628,30, 34,35,40 six studies assessed only ESCC,21,23,25,3638 and seven studies included both types.1821,27,35,37
All 21 studies assessed PR, of which 9 studies used the Mandard TRG scoring system,23,24,2628,30,32, 33,36 9 studies only assessed whether pCR was achieved21,22,25,31,34,35, 37,38,41 (defined as no viable tumor cells; ypT0), one study used the assessment of the College of American Pathologist (1–3),29 1 study used pathologic grading according to Schneider et al.,39,42 and 1 study used pathologic grading according to Chirieac et al.43 Eight studies assessed immune markers, of which PD-L1, PD-1, CD80, CD8, CD4 and CD3 were assessed most extensively.2330 Three studies assessed CD8, PD-L1 and PD-1 in diagnostic tumor biopsies before nCRT,24,27,28 while six studies assessed CD4, CD8, PD-L1, PD-1, CD80 and CD3 in surgical resection specimens after nCRT.2326,29,30
Two studies determined whether diabetes mellitus (DM) affected pathologic outcome.31,41 In one of these studies, diabetic and non-diabetic patients were matched on patient and tumor characteristics.31 Both studies included both type 1 and type 2 DM.
All studies on CR included a baseline and post-nCRT 18F-FDG-PET scan. Eleven studies assessed CR,21,22,3240 of which six studies assessed ΔSUVmax,21,32,34,35,38, 40 four studies assessed percentage reduction SUVmax,34, 3739 four studies assessed mean tumor volume (MTV),32, 33,36,40 two studies assessed tumor lesion glycolysis (TLG),36,40 and three studies assessed ΔSUVmean.3234

Effect of Metabolic Markers on Pathologic Response

Two studies on the effect of DM on pathologic response were included and are shown in Table 2. In total, 73 diabetic patients and 293 non-diabetic patients were included. DM was associated with a decreased likelihood of achieving pCR according to Alvarado et al.,31 whereas Boyd et al. showed no significant difference between both groups.41
Table 2
Effect of metabolic marker diabetes on pathologic response (no 18F-FDG-PET/CT)
Author
Pathologic response
Diabetes (%)
No diabetes (%)
Total (%)
p-Value
Alvarado et al., 202231 a
    
0.03
 
TRG1
5 (9.3)
41 (21.6)
46 (18.9)
 
 
TRG2–5
49 (90.7)
149 (78.4)
198 (81.1)
 
Total
 
54 (100.0)
190 (100)
244 (100)
 
Boyd et al., 201041 a
    
0.62
 
TRG1
7 (36.8)
32 (31.1)
39 (10.7)
 
 
TRG2–5
12 (63.2)
71 (68.9)
83 (22.7)
 
Total
 
19 (100.0)
103 (100.0)
122 (33.3)
 
Bold value indicates the significant values (p < 0.05)
a Univariate and multivariate regression
TRG tumor regression grade according to Mandard, 18F-FDG-PET/CT F-18 fluorodeoxyglucose positron emission tomography/computed tomography

Effect of Immune Markers on Pathologic Response

Tables 3 and 4 show the pathologic immune markers on PR in treatment-naïve biopsies (Table 3) and surgical resections after nCRT (Table 4) in the primary tumor/TME/overall tumor area. Treatment-naïve biopsies were collected and assessed on immune markers prior to nCRT. The median density of immune markers was assessed in the total area.
Table 3
Effect of immune markers on pathologic response in the total area, tumor sample, and tumor microenvironment of treatment-naïve biopsies (no 18F-FDG-PET/CT)
Pre-CRT
Author
Assessment
Cut-off value
Sample size
n pGR (TRG1–3)
n pPR (TRG4–5)
Correlation biomarkers with PR in biopsies
p-Value total area
p-Value tumor
p-Value TME
CD8
          
 
Gobel et al., 202228 a,b
Cell count/mm2
Median
58
47
11
No significant difference in CD8 density between pGR and pPR
0.223
NA
NA
 
Goedegebuure et al., 202124 a,b
Cell count/mm2
Median
40
31
9
High intratumoral density of CD8+ is associated with pGR
No significant difference in stromal density between groups
NA
0.031
NM
 
Soeratram et al., 202127 c
Cells/mm2
Mean and median
81
48
33
Higher combined mean density of CD8 was associated with pGR
0.001
0.013
0.026
PD-1
          
 
Gobel et al., 202228 a,b
<1%, 1 to <10%, 10 to <50%, ≥50%
58
47
11
Intra- and peritumoral PD-1 had no significant influence on TRG
NA
NM
NM
 
Soeratram et al., 202128 c
cells/mm2
Mean and median
81
48
33
pPR had significant higher stromal PD-1 in lymphocytes compared with pGR
NA
0.222
0.048
PD-L1
          
 
Gobel et al., 202228  a,b
<1%, 1 to <10%, 10 to <50%, ≥50%
58
47
11
PD-L1-positive score in the tumoral area: no significant influence; PD-L1-positive score in the peritumoral area was found significantly more in pGR
NA
NM
0.036
 
Soeratram et al., 202127  c
<1–4%, 5–24%, 25–100%
Mean and median
81
48
33
CPS > 1 was associated with lower TRG (1–3)
0.010
NA
NA
Bold values indicate the significant values (p < 0.05)
a Pearson’s Chi-square test
b Two-tailed z-test
c Mann–Whitney U-test
18F-FDG-PET/CT F-18 fluorodeoxyglucose positron emission tomography/computed tomography, TRG tumor regression grade, pGR pathologic good responders, pPR pathologic poor responders, PR pathologic response, CRT chemoradiotherapy, CPS combined positive score, PD-1 programmed death-1, PD-L1 programmed death-ligand 1, TME tumor microenvironment, NA not available, NM not mentioned
Table 4
Effect of immune markers on pathologic response in the total area, tumor sample, and tumor microenvironment of surgical specimens (no 18F-FDG-PET/CT)
Post-CRT author
Assessment
Cut-off value
Sample size
n pGR (TRG1–2)
n pPR (TRG3–5)
Correlation biomarkers with PR in surgical specimens
p-Value total area
p-Value tumor
p-Value TME
CD8
         
Fassan et al., 201944 a,b
Cell count/5 HPF
Median
88
23
65
pGR had significant enrichment of CD8+ compared with pPR
<0.001
NA
NA
Goedegebuure et al., 202124 a
Cell count/mm2
Median
40
12 (TRG1)
28 (TRG2–5)
TRG1 was significantly more present in a CD8-dominant infiltrate
0.027
NA
NA
Haddad et al., 202246 c
% ± SD
Mean
17
11
6
Significant enrichment of CD8+ in pGR compared with pPR
NA
<0.001
0.001
Koemans et al., 202130 c
Cell count/mm2
4 hotspots of 0.5 mm*0.5mm2
123
62
61
pGR had significantly less CD8+ compared with pPR
0.001
NA
NA
Kotsafti et al., 202126 a,b
Cell count/5 HPF
Median
123
20
103
Peritumoral healthy mucosa in pGR had significant high CD8+ compared with pPR
0.05
NA
NA
Soeratram et al., 202127 a
Cells/mm2
Mean and median
89
55 (TRG1–3)
25 (TRG4–5)
Higher CD8 in the tumor epithelium was associated with pPR; higher CD8 in tumor stroma was associated with pGR
NA
0.000
0.000
CD4
         
Fassan et al., 202244 a,b
Cell count/5 HPF
Median
88
23
65
pGR had significant enrichment of CD4+ compared with pPR
0.006
NA
NA
Haddad et al., 202246 c
% ± SD
Mean
28
13
15
pGR had significant enrichment of CD4+ compared with pPR
NA
0.009
0.004
Koemans et al., 202130 c
Cell count/mm2
4 hotspots of 0.5 mm*0.5mm2
123
62
61
pGR had significantly less CD4+ compared with pPR
<0.001
NA
NA
CD3
         
Haddad et al., 202246 c
% ± SD
Mean
88
13
15
pGR had significant enrichment of CD3+ compared with pPR
NA
<0.001
<0.001
Koemans et al., 202130 c
Cell count/mm2
4 hotspots of 0.5 mm*0.5mm2
123
62
61
pGR had significant less CD3+ compared with pPR
<0.001
NA
NA
CD80
         
Fassan et al., 202244 a,b
Cell count/5 HPF
Median
88
23
65
No difference between pGR and pPR
0.4874
NA
NA
Kotsafti et al., 202126 a,b
Cell count/5 HPF
Median
123
20
103
No difference between pGR and pPR
0.89
NA
NA
PD-1
         
Kotsafti et al., 202126 a,b
NA
123
20
103
pGR had significantly lower mRNA PD-1 compared with pPR
0.0065
NA
NA
PD-L1
         
Fassan et al., 201944 a,b
Cell count/5 HPF
88
23
65
pGR had significant levels of PD-L1 expressions either on tumor cells or in lymphocytes than pPR
NA
0.004
NA
Huang et al., 201925 d
>1% = positive, <1 = negative
107
28
79
Positive tumoral PD-L1 expression was significantly associated with pPR
NA
0.036
NA
Koemans et al., 202130 c
0%, 1–5%, 6–9%, 10–29%, >30%
123
62
61
No association between PD-L1 positivity in tumor cells and PR; pPR had significantly more PD-L1-positive lymphocytes
NA
1.00
0.001
Kotsafti et al., 202126 a,b
NA
123
20
103
pGR had significantly lower mRNA PD-L1 compared with pPR
0.0005
NA
NA
Soeratram et al., 202127 a
<1–4%, 5–24%, 25–100%
Mean and median
89
55 (TRG1–3)
25 (TRG4–5)
CPS > 1 was associated with pPR
0.010
NA
NA
Bold values indicate the significant values (p < 0.05)
a Pearson’s Chi-square test
b Two-tailed z-test
c Mann–Whitney U-test
d Logistic regression
18F-FDG-PET/CT F-18 fluorodeoxyglucose positron emission tomography/computed tomography, TRG tumor regression grade, pGR pathologic good responders, pPR pathologic poor responders, PR pathologic response, CRT chemoradiotherapy, CPS combined positive score, TME tumor microenvironment, NA not available, PD-1 programmed death-1, PD-L1 programmed death-ligand 1, HPF high power field, SD standard deviation
As the included studies combined different TRG groups, we were unable to create consistent TRG groups for this review. Tumor regression in these studies was based on vital tumor tissue at the ratio of fibrosis. In addition, patients with pCR (TRG1) were considered free of residual tumor, which is less likely compared with those with non-pCR (TRG2–5). Therefore treatment-naïve biopsies (Table 3) were divided according to the pathologic examination of the resected specimen in good (TRG1–3) and poor (TRG4–5) responders. The Mandard response rates from the treatment-naïve biopsies were extrapolated from their resected specimens. In assessing potential biomarkers in the resected specimen (Table 4), responders after nCRT were divided into pathologic good responders (TRG1–2) and pathologic poor responders (TRG3–5).
Table 3 shows that an overall higher tumoral and TME infiltration of CD8 in treatment-naïve biopsies was associated with a better PR (p = 0.013 and p = 0.026; p = 0.001; p = 0.031, respectively)24,27 Moreover, a higher PD-1 in the TME seemed to significantly predict the possible poor response in tumor tissue from treatment-naïve biopsies (p = 0.048) (Table 4); however, PD-1 in the primary tumor was shown to not be predictive for tumor response (p = 0.222) (Table 3).27,28 PD-L1 expression in the treatment-naïve biopsies showed to predict better PR (lower TRG) both in the TME as the overall tumoral and the TME area (p = 0.036, p = 0.010, respectively).25,27 Only Huang et al. showed that a high density of PD-L1 in the treatment-naïve biopsies predicted poor PR (higher TRG) (p = 0.036).25
Table 4 shows that tumoral and stromal CD8 was found to be significantly higher in pathologic good responders as well as in the healthy mucosa in resected specimens after nCRT.26,4446 Soeratram et al., who distinguished tumoral and stromal CD8, showed that stromal CD8 was significantly associated with good pathologic response (p = 0.000, whereas tumoral CD8 was correlated with a poorer pathologic response (p = 0.000).27 Koemans et al. showed that good responders had significantly less CD8 in the overall area compared with poor responders after nCRT (p = 0.001).30
The majority of the studies found significant enrichment of CD4 in the tumor and the TME in surgical resection specimens after CRT (p = 0.006, p = 0.009, p = 0.004, respectively) (Table 4);23,29 however, one study contradicted these results and showed that poor responders had significant enrichment of CD4 density compared with poor responders (p ≤ 0.001).30
Furthermore, higher PD-1 in the overall tumor and stromal area was shown to be significantly predictive for a poor PR after nCRT (p = 0.0065).26
PD-L1 expression after nCRT proved to be associated with a poor PR according to Koemans et al. (p = 0.001).30 Moreover, a high PD-L1 in the overall area was correlated with a poor PR after nCRT (p = 0.0005, p = 0.010, respectively).26,27
Regarding CD80, two studies revealed no differences in CD80 between pathologic good and poor responders in the overall tumoral and stromal area after nCRT (p = 0.4874, p = 0.89, respectively).26,44

Effect of Clinical Metabolic Markers on Pathological Response

We considered the semi-quantative tools that are used for measuring glucose metabolism and 18F-FDG uptake (SUVmax, SUVmean, ΔSUVmax and percentage reduction SUVmax) in the 18F-FDG-PET/CT scan as clinical metabolic markers. Table 5 describes the effect of ΔSUVmax, percentage reduction SUVmax, ΔSUVmean, TLG, MTV, and ΔSUVratio on pathologic response. Pathologic responders were divided into good responders (TRG1–2) and poor responders (TRG3–5).
Table 5
Effect of immune and metabolic markers on pathologic response (in the presence of 18F-FDG-PET/CT)
 
Author
Total sample size
n pGR (TRG1–2)a
n pPR (TRG3–5)a
Correlation clinical biomarkers with pathologic tumor response
p-Value
ΔSUVmax
      
 
Fang et al., 201732 b
20
14
6
ΔSUVmax was not correlated with pCR on interim and post-CRT scan
0.508;
1.00
Kukar et al., 201534 c
77
22
55
ΔSUVmax was higher in pGR
0.03
Arnett et al., 201635 d
193
NM
NM
ΔSUVmax was not correlated with pCR
0.25
Lee et al., 202138 e
158
44
114
ΔSUVmax did not differ between pCR and pPR
0.201
Van Rossum et al., 201740 d
70
27
43
Higher ΔSUVmax was significantly related to a good response
0.01
Li et al., 202121 d
127
57 (TRG1)
70 (TRG2–5)
ΔSUVmax was an independent predictor for pCR
0.002
% reduction in SUVmax
      
Kukar et al., 201534 c
77
22
55
% reduction SUVmax was higher in pGR
0.03
Lee et al., 202138 e
158
44
114
% reduction SUVmax did not differ between pCR and pPR
0.071
Piessen et al., 201339 b
60
21
25
No significant difference in % reduction SUVmax between pGR and pPR
0.310
Dewan et al., 201737 d
70
24 (TRG1)
46 (TRG2–5)
% reduction SUVmax of 72.32% predicts pCR (sensitivity 70.8%, specificity 67.4%)
0.011
TLG
      
Choi et al., 202136 d
275
75 (TRG1)
200 (TRG2–5)
Higher pre-CRT TLG (> 205.67) was associated with a lower probability of pCR
0.0318
Van Rossum et al., 201740 d
70
27
43
Higher post-TLG was associated with a higher chance of pPR
0.01
MTV
      
Fang et al., 201732 b
20
14
6
MTV was not correlated with pCR on interim and post-CRT scan
0.198; 0.6
Gillham et al., 200633 c
32
9
23
No correlation between MTV and TRG
0.472
Choi et al., 202136 d
275
75 (TRG1)
200 (TRG2–5)
Higher post-MTV (> 4.99) was associated with a low probability of pCR
0.0005
Van Rossum et al., 201740 d
70
27
43
Higher post-MTV was associated with a higher chance of pPR
0.01
ΔSUVmean
      
Fang et al., 201732 b
20
14
6
No correlation between ΔSUVmean and TRG on interim and post-CRT scan
0.424; 0.704
Gillham et al., 200633 c
32
9
23
No correlation between ΔSUVmean and TRG
0.645
Kukar et al., 201534 c
77
22
55
ΔSUVmean was higher in pGR
0.03
ΔSUVratio
      
Li et al., 202121 d
127
57 (TRG1)
70 (TRG2–5)
ΔSUVratio was an independent predictor for pCR
0.007
Obesity
      
Wang et al., 201022 d
405
85 (TRG1)
121 (TRG2–5)
BMI is not a significant predictor for pCR
0.9879
Bold values indicate the significant values (p < 0.05)
a In case studies that made different divisions in pathologic responders, the numbers and specific tumor regression grade were indicated in the table
b Mann–Whitney test
c Wilcoxon rank-sum test and Kruskal–Wallis
d Logistic regression
e Student’s t-test
18F-FDG-PET/CT F-18 fluorodeoxyglucose positron emission tomography/computed tomography, pGR pathologic good responders, pPR pathologic poor responders, TRG tumor regression grade according to Mandard, SUVmax maximum standardized uptake value, SUVmean mean standardized uptake value, TLG total lesion glycolysis, MTV metabolic tumor volume, SUVratio standardized uptake value ratio, NM not mentioned, pCR pathologic complete response, BMI body mass index
ΔSUVmax was evaluated in six studies.21,32,34, 35,38,40 Kukar et al. and van Rossum et al. showed that ΔSUVmax was higher in pathologic good responders (p = 0.03, p = 0.01, respectively).34,40 Moreover, Li et al. assessed ΔSUVmax as an independent predictor for pCR (p = 0.002).21 However, Arnett et al. and Lee et al. found no significant difference between ΔSUVmax in good and poor responders.38,47
Four studies assessed the effect of percentage reduction SUVmax,34,3739 of which two showed no significant difference between pathologic good and poor responders.38,48 Kukar et al. showed that pathologic good responders had a higher percentage reduction SUVmax,34 while Dewan et al. set a cut-off of 72.32% reduction of SUVmax to be predictive for pCR.37
TLG was evaluated in two studies, showing that a high TLG before and after CRT was associated with poor PR (p = 0.0318, p = 0.01, respectively).36,40
Four studies assessed the effect of MTV,32,33,36,40 of which two showed that a high post-CRT MTV was correlated with a poor PR (p = 0.0005, p = 0.01, respectively).36,40 The other two studies showed no correlation with PR (p = 0.6, p = 0.472, respectively).32,33
ΔSUVmean was assessed in three studies, of which two showed no correlation between pathologic response.3234 However, Kukar et al. assessed that pathologic good responders had a higher ΔSUVmean compared with poor responders (p = 0.03).34
Only one study evaluated body mass index on PR, which showed no significant prediction for pCR (p = 0.9879).22

Effect of Metabolic and Immune Markers on Clinical Response and Pathologic Response (18F-FDG-PET/CT)

ESM Table 2 shows the effect of immune and metabolic markers on PR and CR. Both studies divided the assessed groups into pCR (TRG1) or no pCR (TRG2–5).
Wang et al. evaluated the effect of obesity as a metabolic marker on CR, which showed not to be a significant predictor (p = 0.46).22 Li et al. assessed the correlation between immune markers neutrophil to lymphocyte ratio (NLR) and PET markers on prediction of PR, which showed that ΔNLR <3 and ΔSUVratio >58% gave the best positive predictive value (84.8%) for pCR.21

Risk-of-Bias Assessment

Risks of bias was assessed for all included studies (n = 21) [ESM Fig. 1]. The individual risk-of-bias scores can be found in ESM Table 3 and ESM Table 4, on each risk of bias for each included study separately.

Discussion

Metabolic and immune biomarkers of the TME have a pivotal role in providing tumor cells the optimal condition to survive and proliferate while also influencing their response to therapy. Due to intratumoral and microenvironmental heterogeneity after nCRT, all available information from the tumor, its TME, and the pathological specimen was included. Here, we provide an overview of potential metabolic and immune TME biomarkers that might play a role in PR and CR after nCRT in EC.
Current research in targeting the metabolic TME is based on 18F-FDG-PET/CT imaging of the altered glycolytic tumor metabolism with acidification of the TME. TME acidification induces hypoxia response pathways and leads to evasion of the immune system, which is associated with high metastatic potential and treatment resistance.49 As such, the upregulation of glycolysis as a measure of extracellular acidification remains a critical step in the activation of immune cells. In this intricate interaction of heterogeneous tumor cells, a variety of secretory cytokines and chemokines from non-malignant cells, i.e., stroma and immune cells, are involved in the efficacy of anticancer therapy. Metabolic remodeling with inflammatory response and oxidative phosphorylation is important in the resistance to neoadjuvant treatment in EC. Recently, a promising novel ex vivo method showed the significance of oxidative phosphorylation in measuring real-time metabolic profiles of treatment-naïve EC biopsies. In clinical imaging of hypoxic response and glycolytic metabolism in malignant tumors, 18F-FDG-PET/CT is most commonly used.50 Based on the assessment of histopathology, the corresponding 18F-FDG-PET/CT response and promising biomarkers markers, nCRT combined with immunotherapy might be considered as an organ-preserving treatment approach in the near future.

Metabolic Tumor Microenvironment (TME) Markers

There were no studies on metabolic TME markers in EC that also assessed the influence of these markers on PR and/or CR after nCRT. Diabetes was suggested as a surrogate metabolic marker. However, the result of this study shows a limited role of DM on PR after nCRT. An overexpression of insulin receptors and insulin-like growth factors lead to the promotion of cell cycle progression and inhibition of apoptosis.51,52 The overexpressed insulin receptors on cancer cells of diabetic patients, who are also characterized by hyperinsulinemia, may be activated, leading to the ability of cancer cells to evade destruction by chemoradiotherapy, resulting in an unfavorable PR and CR.53 As a result, hypoxia and hyperglycemia occur, which might help remodeling the TME into an even more aggressive environment, leading to poorer response to nCRT.54

Immune TME Biomarkers

We showed that high CD3 and CD4 infiltration were generally correlated with better PR. Even though some studies showed no significant difference in CD8 between good and poor pathologic responders, CD8 infiltration in treatment-naïve biopsies was generally significantly associated with a better PR.24,27 One study showed that nCRT was useful to induce CD4 and CD8 infiltration within the TME, suggesting that an elevated level of lymphocytes before nCRT might be a surrogate of a strong immune response induced by tumor cell necrosis caused by chemotherapy.55
The activation of CD8 cells after nCRT might be impaired by persistent high expression of the CXCL12/CXCR4 axis in EC stem cells resulting in a downregulation of major histocompatibility complex (MHC) class I molecules and upregulating immunosuppressive cytokines.56 nCRT can also cause inflammation, leading to an influx of CD8 immune cells. These patients could benefit from the upcoming immune-directed treatment strategies such as PD-1/PD-L1 blockade.5760 In this study, CD8 pre/post-nCRT and CD3/CD4 after nCRT seem to be involved in the antitumor response. Moreover, the location (i.e., tumoral or stromal) at which the CD8 influx occurs might affect active immune behavior. The extracellular matrix or other immune-suppressive cells within the tumor and the TME might barricade the function of tumoral CD8,61,62 resulting in an inefficient function of CD8 intratumorally.
The potential clinical value of tumoral PD-L1 expression in EC patients with residual disease after nCRT with surgery has shown to be significant for DFS after adjuvant anti-PD-1 nivolumab in the Checkmate-577 study, and showed a better PR in the Keynote-590 study with anti-PD-1 pembrolizumab and nCRT.3,7 The included studies also showed that a high proportion of PD-L1 in positive treatment-naïve tumor samples may affect PR; however, the exact mechanism behind this is still unknown. PD-L1 expression in pretreatment biopsies might be different due to intratumoral heterogeneity of EC, in which PD-L1 expression can only be partially captured. However, further investigation is needed.
We also showed that a higher expression of tumoral or stromal PD-L1 after nCRT is generally associated with a poor PR to chemoradiotherapy. Therefore, PD-L1 might be a potential target in EC patients receiving nCRT in order to improve therapy response. Together with the other predictive immune biomarkers, PD-L1 expression in the tumor and its microenvironment could be used to define EC patients with major or poor pathologic response after nCRT with resection and/or a clinical prognostic high- versus low-risk profile. PD-L1 positivity can be expressed by using both the tumor cell (TC ≥ 1% in at least 100 tumor cells in the PD-L1-stained slide) and combined positivity score (CPS ≥10 PD-L1-stained cells, including tumor cells, lymphocytes, macrophages in the associated infiltration). Based on the histologic EC subtypes, these clinical prognostic risk biomarkers and the different predictive response biomarkers between tumor-naïve biopsies and the resected residual tumor material potential biomarkers may be identified for the ypCR and non-ypCR groups.

18F-FDG-PET/CT Biomarkers

An 18F-FDG-PET/CT scan is commonly used in EC patients undergoing the CROSS regimen, to monitor treatment response. Many studies aimed to find a correlation between the semi-quantitative parameters of 18F-FDG-PET/CT with PR. However, our included studies showed contradictory evidence for the value of parameters such as SUV, ΔSUVmax and SUVmax in predicting PR and CR.
A low SUV might be associated with hypoxic tumors, as is the case in EC. An hypoxic environment could emerge if the tumor became more resistant to chemoradiotherapy, leading to a poor pathologic response.63 Moreover, a wide heterogeneity between studies could account for contradictory results, such as different methods and experience at performing and interpreting 18F-FDG-PET/CT scans, methods to calculate PET parameters, physiological factors that may affect SUV uptake (i.e., inflammation) to the esophageal mucosa, scanner technology, chemoradiotherapy schedules, sample size, and methods of data collection. Studies also vary regarding the time interval of post-treatment 18F-FDG-PET/CT after completion of nCRT, which may affect the interpretation of predictive accuracy.
Therefore, the predictive value of other clinical 18F-FDG-PET/CT-based markers needs to be explored. We showed that TLG and MTV might have more potential to predict pathological and clinical outcome, which is also in line with recent studies.64,65 These volume-based 18F-FDG-PET parameters might provide more valuable information that supplement SUV uptake for predicting PR and CR. Future studies should thus focus on combining these parameters and find a clear cut-off value.
The present study has some limitations. Treatment-naïve EC biopsies contain a highly heterogenous inflammatory secretion profile. It is plausible that pretreatment-naïve biopsies are not representative enough. Therefore, it is important to know which specimen has been used in determining the predictive role of biomarkers. First, tumor heterogeneity may be missed in these small standard diagnostic tumor biopsies, and second, we should be aware of changes in biomarkers during chemotherapy and/or radiotherapy.66 Furthermore, biology from resected tissue alone may not reflect tumor biology at diagnosis. Moreover, patients attaining pCR (ypT0/N0) who commonly exhibit a good prognosis will not likely receive adjuvant therapy. Furthermore, we included articles of various markers that were assessed in different ways, i.e., mRNA expression of assessed markers, assessments conducted in healthy esophageal mucosa, and overall density of assessed markers. These differences in assessing various markers made it difficult to interpret the results.

Conclusion

Our systematic review showed that CD8, CD4, CD3, and PD-L1 are promising immune markers in predicting PR. Moreover, we showed that TLG and MTV have potential in predicting CR and PR. Additional research should focus more on combining histopathology and nuclear imaging features in EC before and after nCRT to assess metabolic and immune TME markers.

Disclosures

Conflicts of interest to disclose that are relevant to this article.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
25.
Zurück zum Zitat Huang W-T, Lu H-I, Wang Y-M, et al. Positive Programmed Cell Death-Ligand 1 Expression Predicts Poor Treatment Outcomes in Esophageal Squamous Cell Carcinoma Patients Receiving Neoadjuvant Chemoradiotherapy. J Clin Med. 2019;8(11). doi:https://doi.org/10.3390/jcm8111864 Huang W-T, Lu H-I, Wang Y-M, et al. Positive Programmed Cell Death-Ligand 1 Expression Predicts Poor Treatment Outcomes in Esophageal Squamous Cell Carcinoma Patients Receiving Neoadjuvant Chemoradiotherapy. J Clin Med. 2019;8(11). doi:https://​doi.​org/​10.​3390/​jcm8111864
30.
Zurück zum Zitat Koemans WJ, Van Dieren JM, Den Van Berg JG, et al. High CD8 tumour-infiltrating lymphocyte density associates with unfavourable prognosis in oesophageal adenocarcinoma following poor response to neoadjuvant chemoradiotherapy. Histopathology. 2021;79:238–51. https://doi.org/10.1111/his.14361.CrossRefPubMed Koemans WJ, Van Dieren JM, Den Van Berg JG, et al. High CD8 tumour-infiltrating lymphocyte density associates with unfavourable prognosis in oesophageal adenocarcinoma following poor response to neoadjuvant chemoradiotherapy. Histopathology. 2021;79:238–51. https://​doi.​org/​10.​1111/​his.​14361.CrossRefPubMed
38.
Zurück zum Zitat Lee J, Choi JY, Lim SW, et al. Utility of positron emission–computed tomography for predicting pathological response in resectable oesophageal squamous cell carcinoma after neoadjuvant chemoradiation. Eur J Cardio-thoracic Surg. 2021;58(5):1019–26. https://doi.org/10.1093/EJCTS/EZAA181.CrossRef Lee J, Choi JY, Lim SW, et al. Utility of positron emission–computed tomography for predicting pathological response in resectable oesophageal squamous cell carcinoma after neoadjuvant chemoradiation. Eur J Cardio-thoracic Surg. 2021;58(5):1019–26. https://​doi.​org/​10.​1093/​EJCTS/​EZAA181.CrossRef
40.
Zurück zum Zitat van Rossum PSN, Fried DV, Zhang L, et al. The value of (18)F-FDG PET before and after induction chemotherapy for the early prediction of a poor pathologic response to subsequent preoperative chemoradiotherapy in oesophageal adenocarcinoma. Eur J Nucl Med Mol Imaging. 2017;44(1):71–80. https://doi.org/10.1007/s00259-016-3478-2.CrossRefPubMed van Rossum PSN, Fried DV, Zhang L, et al. The value of (18)F-FDG PET before and after induction chemotherapy for the early prediction of a poor pathologic response to subsequent preoperative chemoradiotherapy in oesophageal adenocarcinoma. Eur J Nucl Med Mol Imaging. 2017;44(1):71–80. https://​doi.​org/​10.​1007/​s00259-016-3478-2.CrossRefPubMed
44.
Zurück zum Zitat Fassan M, Cavallin F, Guzzardo V, et al. CD4+ and CD8+ lymphocyte rate and PDL-1 leukocyte expression are predictive of clinical complete response after neoadjuvant chemoradiotherapy for squamous cell cancer of the thoracic oesophagus. United Eur Gastroenterol J. 2017;5(5):A589. https://doi.org/10.1177/2050640617725676.CrossRef Fassan M, Cavallin F, Guzzardo V, et al. CD4+ and CD8+ lymphocyte rate and PDL-1 leukocyte expression are predictive of clinical complete response after neoadjuvant chemoradiotherapy for squamous cell cancer of the thoracic oesophagus. United Eur Gastroenterol J. 2017;5(5):A589. https://​doi.​org/​10.​1177/​2050640617725676​.CrossRef
48.
Zurück zum Zitat Piessen G, Robb WB, Petyt G, et al. Ineffectiveness of 18f-fluorodeoxyglucose positron emission tomography in the evaluation of tumour response after completion of neoadjuvant chemoradiation in oesophageal cancer. Eur Surg Res. 2013;50(2):195–6. https://doi.org/10.1159/000351963.CrossRef Piessen G, Robb WB, Petyt G, et al. Ineffectiveness of 18f-fluorodeoxyglucose positron emission tomography in the evaluation of tumour response after completion of neoadjuvant chemoradiation in oesophageal cancer. Eur Surg Res. 2013;50(2):195–6. https://​doi.​org/​10.​1159/​000351963.CrossRef
53.
Zurück zum Zitat Armengol-Alonso A, Tusquets I, Servitja S, et al. Association between insulin resistance (Ir) and pathologic complete response (Pcr) to neoadjuvant chemotherapy (Nac) among no diabetic women with localized infiltrating breast cancer (Libc). Ann Oncol. 2014;25(Suppl 4):iv114. https://doi.org/10.1093/annonc/mdu328.13.CrossRef Armengol-Alonso A, Tusquets I, Servitja S, et al. Association between insulin resistance (Ir) and pathologic complete response (Pcr) to neoadjuvant chemotherapy (Nac) among no diabetic women with localized infiltrating breast cancer (Libc). Ann Oncol. 2014;25(Suppl 4):iv114. https://​doi.​org/​10.​1093/​annonc/​mdu328.​13.CrossRef
58.
Zurück zum Zitat Kang YK, Boku N, Satoh T, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390(10111):2461–71. https://doi.org/10.1016/S0140-6736(17)31827-5.CrossRefPubMed Kang YK, Boku N, Satoh T, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390(10111):2461–71. https://​doi.​org/​10.​1016/​S0140-6736(17)31827-5.CrossRefPubMed
Metadaten
Titel
Potential Predictive Immune and Metabolic Biomarkers of Tumor Microenvironment Regarding Pathological and Clinical Response in Esophageal Cancer After Neoadjuvant Chemoradiotherapy: A Systematic Review
verfasst von
H. H. Wang, BSc
E. N. Steffens, BSc
G. Kats-Ugurlu, MD, PhD
B. van Etten, MD, PhD
J. G. M. Burgerhof, MSc
G. A. P. Hospers, MD, PhD
J. T. M. Plukker, MD, PhD
Publikationsdatum
30.09.2023
Verlag
Springer International Publishing
Erschienen in
Annals of Surgical Oncology / Ausgabe 1/2024
Print ISSN: 1068-9265
Elektronische ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-023-14352-z

Weitere Artikel der Ausgabe 1/2024

Annals of Surgical Oncology 1/2024 Zur Ausgabe

Häusliche Gewalt in der orthopädischen Notaufnahme oft nicht erkannt

28.05.2024 Häusliche Gewalt Nachrichten

In der Notaufnahme wird die Chance, Opfer von häuslicher Gewalt zu identifizieren, von Orthopäden und Orthopädinnen offenbar zu wenig genutzt. Darauf deuten die Ergebnisse einer Fragebogenstudie an der Sahlgrenska-Universität in Schweden hin.

Fehlerkultur in der Medizin – Offenheit zählt!

28.05.2024 Fehlerkultur Podcast

Darüber reden und aus Fehlern lernen, sollte das Motto in der Medizin lauten. Und zwar nicht nur im Sinne der Patientensicherheit. Eine negative Fehlerkultur kann auch die Behandelnden ernsthaft krank machen, warnt Prof. Dr. Reinhard Strametz. Ein Plädoyer und ein Leitfaden für den offenen Umgang mit kritischen Ereignissen in Medizin und Pflege.

Mehr Frauen im OP – weniger postoperative Komplikationen

21.05.2024 Allgemeine Chirurgie Nachrichten

Ein Frauenanteil von mindestens einem Drittel im ärztlichen Op.-Team war in einer großen retrospektiven Studie aus Kanada mit einer signifikanten Reduktion der postoperativen Morbidität assoziiert.

TAVI versus Klappenchirurgie: Neue Vergleichsstudie sorgt für Erstaunen

21.05.2024 TAVI Nachrichten

Bei schwerer Aortenstenose und obstruktiver KHK empfehlen die Leitlinien derzeit eine chirurgische Kombi-Behandlung aus Klappenersatz plus Bypass-OP. Diese Empfehlung wird allerdings jetzt durch eine aktuelle Studie infrage gestellt – mit überraschender Deutlichkeit.

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.