Skip to main content
Erschienen in: BMC Cancer 1/2020

Open Access 01.12.2020 | Research article

Worse treatment response to neoadjuvant chemoradiotherapy in young patients with locally advanced rectal cancer

verfasst von: Yiyi Zhang, Liangliang Yan, Yong Wu, Meifang Xu, Xing Liu, Guoxian Guan

Erschienen in: BMC Cancer | Ausgabe 1/2020

Abstract

Background

To evaluate the impact of age on the efficacy of neoadjuvant chemoradiotherapy (NCRT) in patients with locally advanced rectal cancer (LARC).

Method

LARC patients undergoing NCRT and radical surgery from 2011 to 2018 were divided into young (< 40 years) and old (≥40 years) groups. Multivariate analyses were performed to identify predictive factors for pathological complete response (pCR). Predictive nomograms and decision curve analysis were used to compare the models including/excluding age groups. Immunohistochemical analysis was performed to detect CD133 expression in LARC patients.

Result

A total of 901 LARC patients were analyzed. The young group was associated with poorly differentiated tumors, more metastatic lymph nodes, higher perineural invasion, and a lower tumor regression grade (P = 0.008; P < 0.001; P < 0.001; P = 0.003). Logistic regression analysis demonstrated that age < 40 years (HR = 2.190, P = 0.044), tumor size (HR = 0.538, P < 0.001), pre-NCRT cN stage (HR = 0.570, P = 0.036), and post-NCRT CEA level (HR = 0.877, P = 0.001) were significantly associated with pCR. Predictive nomograms and decision curve analysis demonstrated that the predictive ability of models including the age group was superior to that of models excluding the age group. Higher CD133 expression was more common in young LARC patients.

Conclusion

Young patients with LARC were associated with lower pCR rates following NCRT. The ability of the predictive model was greater when based on the age group. Young LARC patients were associated with a higher CD133+ tumor stem cell burden, which contributed to the lower pCR rates.
Hinweise
Yiyi Zhang, Liangliang Yan, and Yong Wu contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
NCRT
Neoadjuvant chemoradiotherapy
LARC
Locally advanced rectal cancer
CRC
Colorectal cancer
TME
Total mesorectal excision
CEA
Carcinoembryonic antigen
MRI
Magnetic resonance imaging
pCR
Pathological complete response
ASA
American society of anesthesiology
CRM
Circumferential resection margin
HR
Hazard ratio
DFS
Disease free survival

Background

Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer-related mortality in the USA [1]. CRC is generally thought to be a malignancy affecting the elderly  patients. Over the last two decades, the incidence of CRC has increased in young individuals, especially those aged under 40. However, most studies focus on older CRC patients, especially the elderly (> 70 years) [2, 3]. Few studies have focused on the impact of young age (< 40 years) in CRC patients. In contrast to CRC in the elderly, young patients present at a more advanced tumor stage, with a more aggressive pathological subtype, and poor prognosis [46]. The increasing prevalence in CRC patients aged < 40 years highlights a genuine need to better understand this disease in such patients.
Neoadjuvant chemoradiotherapy (NCRT) and radical resection have become the standard treatment for patients with locally advanced rectal cancer (LARC). The well-documented benefits of this multimodality include a higher probability of tumor downsizing and downstaging, improved tumor resectability and sphincter preservation, and better local tumor control [79]. Approximately 10–30% of patients will achieve a pathological complete response (pCR), heralding an excellent prognosis due to low rates of local and distant recurrence [7, 10, 11]. Major efforts have been devoted to evaluating the influence of age on the efficacy and compliance of NCRT. While most studies were focused on old patients, few studies have focused on young LARC patients. Considering the aggressive tumor biology in young patients, we hypothesize that the efficacy of NCRT is reduced in young patients. Several recent studies have evaluated the impact of age on the efficacy of adjuvant chemotherapy in patients with rectal cancer [2, 12, 13]. Cancer stem cells (CSCs) have the capacity for multipotency and self-renewal and may be responsible for neoplasm formation, metastasis, recurrence, and therapeutic resistance [1416]. Thus, we hypothesize that the early onset is due to higher malignancy cells, such as CSCs in young patients. Moreover, CSCs also indicate a poorer response to NCRT in LARC patients. However, data regarding the association of young age with treatment response to NCRT are scarce, and the influence of young age on the efficacy of NCRT in LARC patients remains unclear.
In this context, the present study was conducted to compare the efficacy of NCRT in young (< 40 years) and old (≥40 years) patients with LARC in terms of tumor response. Additionally, we further investigated the relationship between young age and CSC (CD133 expression) frequency in LARC patients following NCRT.

Patients and method

Patient eligibility

A retrospective study based on our prospectively maintained database was performed. LARC patients who underwent NCRT and radical resection between 2011 and 2018 were identified. The inclusion criteria were as follows: 1) clinical stage II or III (cT3/4 or cN1/2) disease; 2) pathologically proven rectal adenocarcinomas; and 3) tumors located within 12 cm from the anal verge. Exclusion criteria included: 1) previous of malignancies or concurrent with malignancies; 2) patients who underwent emergent surgery, palliative resection, or local excision. Finally, a total of 901 LARC patients were included. Since the majority of patients were pathologically confirmed by endoscopic biopsy when admitted to our hospital (a tertiary referral hospital), colonoscopy samples from only 169 patients were available for the immunohistochemical analysis. This study was approved by the Institutional Review Board (IRB) of Fujian Medical University Union Hospital. A patient flow diagram was shown in Fig 1.

Treatment protocol

Comprehensive assessments for tumor staging of patients were performed by a digital rectal examination, colonoscopy, chest radiography, abdominopelvic magnetic resonance imaging (MRI), and/or transrectal ultrasound (ERUS). Preoperative long-course radiotherapy consisted of a total dose of 45 Gy to the pelvis, delivered in 25 fractions for 5 consecutive weeks (180 cGy per fraction, 5 days a week), followed by a boost of 5.4 Gy to the primary tumor. Preoperative concurrent chemotherapy was initiated on the first day of radiotherapy by using 5-FU plus oxaliplatin (FOLFOX) or capecitabine plus oxaliplatin (CapeOX).
Surgical operation was performed at an interval of 6–8 weeks after the completion of radiation. Surgical resection for rectal cancer was performed according to the principle of total mesorectal excision (TME) and high ligation of the inferior mesenteric artery. The surgical procedure consisted of low anterior resection (LAR), abdominoperineal resection (APR), or Hartmann’s procedure. About 3–4 weeks after surgery, postoperative adjuvant chemotherapy was administered to patients (using FOLFOX or CapeOX) for 6 months.

Definitions

Tumor response to NCRT was graded according to the tumor regression grade (TRG) system [17]. pCR was defined as the absence of viable tumor cells in either the primary tumor site or the resected lymph nodes. Postoperative morbidity was classified according to the Clavien-Dindo classification, grades I-II was considered as minor complications, and grades III-V as major complications. Tumor size was divided according to the quartile intervals to make it more applicable in clinical practice (≤1.8 cm, 1.9–3.1 cm, ≥3.2 cm). Perioperative mortality was defined as any death within 30 days of surgery or occurring in the hospital.

Immunohistochemical analysis

CD133 (Affinity Biosciences, AF5120, Polyclonal, 1:100) protein expression in specimens obtained before NCRT in 169 LARC patients was measured using the immunohistochemical streptavidin-biotin complex method (Fig. 3e, f, g, h) [18]. Phosphate-buffered saline (PBS) was used as the negative control and the image of the positive control from GE Healthcare Life Sciences. Immunoreactivity was scored by semi-quantitative analysis, and the fields were randomly selected in five directions (up, center, down, left, and right) under high magnification (× 400). The color was determined based on the intensity score as follows: 0 (no staining), 1 (light yellow), 2 (brown), and 3 (deep brown). The percentage of positive cells was scored as 0 (< 5%), 1 (5–25%), 2 (25–50%), 3 (50–75%), and 4 (> 75%). The mean value was calculated for each case with the aforementioned scoring methods and the final score was obtained by multiplying these two scores. All analyses were performed in a double-blinded manner.

Statistical analysis

Statistical analysis was performed using SPSS version 23.0 (SPSS INC., Chicago, USA) and R software packages, version 3.5.1 (The R Foundation for Statistical Computing, http://​www.​r-project.​org/​). The categorical variables were presented as frequencies and percentages and assessed using the Chi-squared (χ2) test or Fisher’s exact test. The continuous variables were described as means ± standard deviation and assessed using Student’s t-test. All significant variables in the univariate analysis were entered into a Logistic regression model to identify predictors of pCR. Based on the multivariable analysis, a predictive nomogram was developed using the R project. The performance of the nomogram was evaluated by calculating the Harrell’s concordance index (c-index). Decision curve analysis (DCA) was performed to evaluate the clinical utility of the model for pCR. DCA is a method for evaluation and comparison of the predictive value between different prediction models [19, 20]; therefore, this method was used to evaluate the clinical utility of the model for pCR. The x-axis of the DCA represents the percentage of threshold probability, and the y-axis represents the net benefit of the predictive model. The net benefit was calculated according to the following formula: Net benefit = (true positives/n) − (false positives/n) * (pt/(1 − pt). “Number high risk” indicated the number of patients classified as positive (high risk) by a model including age group according to various threshold probabilities. “Number high risk with the event” was the true positive patient number according to various threshold probabilities. The optimal cut-off values for CD133 expression were calculated and determined by using the X-tile program (http://​www.​tissuearray.​org/​rimmlab/​), a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization, which identified the cut-off with the minimum p values from log-rank χ2 statistics in terms of OS [21]. The optimal cut-off score was identified as 11 for CD133. Thus, we defined CD133 expression score ≤ 11 as low expression, and > 11 as high expression. Survival outcomes were assessed using the Kaplan–Meier method and log-rank test. P < 0.05 was considered to indicate statistical significance.

Results

Patient characteristics

A total of 901 LARC patients were eligible for our analysis. Among them, 75 (8.3%) were assigned to the young group and 826 (91.7%) patients were assigned to the old groups. The median ages in the two groups were 34.3 and 58.1 years, respectively. Additionally, the old group was associated with a higher American Society of Anaesthesiology (ASA) grade (P < 0.05). As shown in Table 1, there were no significant differences between the two groups in terms of sex, the interval between NCRT and surgery, distance from the anal verge, clinical T stage, clinical N stage, pre-NCRT CA19–9 level, pre-NCRT CEA level, post-NCRT CA19–9 level, and post-NCRT CEA level (all P > 0.05),
Table 1
Patient characteristics in patients with LARC after NCRT
Characteristics
Young group (n = 75)
Old group (n = 826)
p value
Sex (%)
  
0.527
 Male
52 (69.3)
537 (65.0)
 
 Female
23 (30.7)
289 (35.0)
 
Age (years)
34.3 ± 4.1
58.1 ± 9.4
< 0.001
ASA score (%)
  
< 0.001
 1
75 (100)
603 (73.0)
 
 2
0
209 (25.3)
 
 3
0
14 (1.6)
 
Distance from the anal verge (cm)
6.4 ± 2.5
6.3 ± 2.3
0.684
Time interval between CRT and surgery (weeks)
9.1 ± 1.4
9.4 ± 2.9
0.468
Clinical T stage (%)
  
0.173
 T1
1 (1.3)
2 (0.2)
 
 T2
0
18 (2.2)
 
 T3
25 (33.3)
317 (38.4)
 
 T4
49 (65.3)
489 (59.7)
 
Clinical N stage (%)
  
1.000
 N0
7 (9.3)
79 (9.5)
 
 N+
68 (90.7)
747 (90.5)
 
Pre-NCRT CEA level (%)
  
0.562
  < 5.0 ng/ml
25(33.3)
283 (34.3)
 
  ≥ 5.0 ng/ml
24(32.0)
219 (26.5)
 
 Unknown
26(34.7)
324 (39.2)
 
Pre-NCRT CA19–9 level (%)
  
0.641
  < 39.0 U/ml
40 (53.3)
430 (52.1)
 
  ≥ 39.0 U/ml
9 (12.0)
76 (9.2)
 
 Unknown
26(34.7)
320 (38.7)
 
Post-NCRT CEA level (%)
  
0.115
  < 5.0 ng/ml
67 (89.3)
672 (81.4)
 
  ≥ 5.0 ng/ml
8 (10.7)
154 (18.6)
 
Post-NCRT CA19–9 level (%)
  
0.320
  < 39.0 U/ml
68 (90.7)
775 (93.8)
 
  ≥ 39.0 U/ml
7 (9.3)
51 (6.2)
 
LARC Locally advanced rectal cancer; NCRT Neoadjuvant chemoradiotherapy; ASA American Society of Anesthesiologists; CRT Chemoradiotherapy; CEA Carcinoembryonic Antigen; CA19–9 Carbohydrate Antigen 19–9

Perioperative, pathological and survival outcomes

No significant differences were observed between the two groups in terms of estimated blood loss, operation time, surgical approach, peri-NCRT complication rates, and sphincter-saving procedure (all P > 0.05, Table 2). There were no significant differences between the two groups in terms of postoperative hospital stay and postoperative complications (P = 0.124, P = 0.736, respectively). Similarly, the chemotherapy regimen did not differ between the two groups (P = 0.461). Compared to the young group, mucinous or signet ring cell carcinoma, (17.3% vs. 7.6%, P = 0.008) or poorly differentiated tumors (24.3% vs. 9.0%, P < 0.001) were more common in the old group. Moreover, the young group was associated with a higher TRG (P = 0.003), as well as a higher rate of perineural invasion (17.3% vs. 6.5%, P = 0.002). Pathological TNM stage and pathological type were similar in both groups (P = 0.957, P = 0.936). Similarly, there were no differences between the two groups in terms of vascular invasion and tumor size did (P = 0.346, P = 0.069). A positive circumferential resection margin (CRM) was observed in one patient (1.3%) in the young group, 10 patients (1.2%) in the old group, with no significant difference between the two groups (P = 1.000). Moreover, Kaplan-Meier curve analysis demonstrated that young patients were associated with poorer prognosis in LARC patients following NCRT. The 3-year OS rate in the old group (≥40 years) was significantly higher than that in the young group (< 40 years) (88.3% vs. 71.6%; P = 0.01, Fig. 3m). Moreover, the 3-year DFS rate for the old group (≥40 years) was higher than that in the young group (< 40 years) (83.8% vs. 68.6%; P = 0.204, Fig. 3l).
Table 2
Operative and postoperative outcomes in patients with LARC after NCRT
Characteristics
Young group (n = 75)
Old group (n = 826)
p value
Operative time (min)
232.5 ± 81.2
225.8 ± 64.0
0.394
Estimated blood loss (ml)
77.7 ± 74.2
88.9 ± 105.5
0.372
Surgery approach (%)
  
0.882
 Laparoscopic
49 (65.3)
549 (66.5)
 
 Open
18 (24.0)
198 (23.9)
 
 Robotic
8 (10.7)
79 (9.6)
 
Postoperative hospital stay (days)
7.8 ± 3.4
8.8 ± 5.5
0.124
Postoperative complications (%)
12 (16.0)
122 (14.9)
0.736
30 days readmission (%)
1 (1.3)
5 (0.6)
0.407
Peri-CRT complicationsa
25 (33.3)
259 (31.4)
0.699
 Major
1 (1.3)
23 (2.8)
0.741
Sphincter-saving procedure (%)
66 (88.0)
721 (87.4)
1.000
Pathological type (%)
  
0.936
 Ulcering
72 (96.0)
796 (96.7)
 
 Expanding
1 (1.3)
11 (1.2)
 
 Infiltrating
2 (2.7)
19 (2.1)
 
Histopathology (%)
  
0.008
 Adenocarcinoma
62 (82.7)
763 (92.4)
 
 Mucinous or signet ring cell carcinoma
13 (17.3)
63 (7.6)
 
Tumor differentiation (%)
  
< 0.001
 Well to moderately differentiated
57 (75.7)
752 (91.0)
 
 Poorly differentiated and others
18 (24.3)
74 (9.0)
 
Chemotherapy regimen (%)
  
0.461
 FOLFOX/CapeOX
33 (44.0)
326 (39.5)
 
 Capecitabine
42 (56.0)
500 (60.5)
 
Lymph nodes retrieved
19.3 ± 13.1
12.0 ± 6.2
< 0.001
Metastatic lymph nodes
2.2 ± 6.1
0.7 ± 2.0
< 0.001
CRM involvement (%)
1 (1.3)
10 (1.2)
1.000
Tumor size (cm)
  
0.069
  ≤ 1.8 cm
17(22.7)
212 (25.7)
 
 1.9–3.1 cm
31 (41.3)
416 (50.4)
 
  ≥ 3.2 cm
27 (36.0)
198 (24.0)
 
Pathological TNM stage (%)
  
0.957
 0
9 (12.0)
183 (22.2)
 
 I
14 (18.7)
205 (24.8)
 
 II
16 (21.3)
217 (26.3)
 
 III
31 (41.3)
179 (21.7)
 
 IV
5 (6.7)
42 (5.2)
 
TRG grade (%)
  
0.003
 0
9 (12.0)
183 (22.2)
 
 1
20 (26.7)
272 (32.9)
 
 2
33 (44.0)
313 (37.9)
 
 3
13 (17.3)
58 (7.0)
 
Perineural invasion (%)
13 (17.3)
54 (6.5)
0.002
Vascular invasion (%)
4 (5.3)
29 (3.5)
0.346
Adjuvant chemotherapy (%)
  
0.484
 Complete
44 (58.7)
427 (51.7)
 
 Decreased complete
9 (12.0)
87 (10.5)
 
 Refuse
2 (2.7)
44 (5.3)
 
 Unknown
20 (26.7)
268 (32.4)
 
a Some patients experienced more than one complication, and categorized as
NCRT Neoadjuvant chemoradiotherapy; CRM Circumferential resection margin; TRG Tumor regression grade

Association between young age and pCR

To explore the association between young age and treatment response to NCRT, we identified predictive factors for pCR by Logistic regression analysis. In the univariate analysis, tumor size (≤1.8 cm vs. 1.9–3.1 cm OR = 6.764, P < 0.001; vs. ≥3.2 cm OR = 2.022, P = 0.007), age (≥40 years vs. < 40 years, OR = 2.087, P = 0.044), pre-NCRT clinical T stage (OR = 0.731, P = 0.031), pre-NCRT clinical N stage (OR = 0.550, P = 0.016), distance from the anal verge (OR = 0.919, P = 0.018), and post-NCRT CEA (OR = 0.381, P < 0.001) were significantly associated with pCR in LARC patients. In the multivariate analysis, tumor size (≤1.8 cm vs. 1.9–3.1 cm OR = 6.764, P < 0.001; vs. ≥3.2 cm OR = 2.022, P = 0.007), age (≥40 years vs. < 40 years, OR = 2.382, P = 0.027), pre-NCRT clinical N stage (OR = 0.560, P = 0.029), and post-NCRT CEA (OR = 0.873, P = 0.001) were independent predictive factors for pCR in LARC patients (Table 3).
Table 3
Univariate and multivariate analysis of predictive factors for pCR in LARC patients (n = 901)
Variables
Univariate analysis
Multivariate analysis
HR
95% CI
p value
HR
95% CI
p value
Sex (male vs. female)
1.351
0.973–1.875
0.073
   
Age (≥40 years vs. < 40 years)
2.087
1.020–4.269
0.044
2.382
1.105–5.137
0.027
ASA
0.964
0.686–1.353
0.831
   
Distance from the anal verge
0.919
0.856–0.986
0.018
0.949
0.878–1.025
0.181
Tumor size
  ≤ 1.8 cm
Reference
Reference
< 0.001
Reference
Reference
< 0.001
 1.9–3.1 cm
6.764
4.019–11.385
< 0.001
5.806
3.412–9.878
< 0.001
  ≥ 3.2 cm
2.022
1.212–3.373
0.007
1.853
1.101–3.119
0.020
Time interval between NCRT and surgery
1.041
0.988–1.097
0.131
   
Pre-NCRT cT stage
0.731
0.553–0.967
0.031
0.816
0.607–1.097
0.177
Pre-NCRT cN stage
0.550
0.338–0.895
0.016
0.560
0.332–0.943
0.029
Post-NCRT CEA level
0.381
0.224–0.647
< 0.001
0.873
0.805–0.946
0.001
Post-NCRT CA19–9 level
0.492
0.219–1.101
0.084
   
Chemotherapy regimen
1.083
0.783–1.499
0.630
   
Radication dose reduction
0.978
0.360–2.653
0.965
   
NCRT complications
0.900
0.641–1.264
0.542
   
pCR Pathological complete response; NCRT Neoadjuvant chemoradiotherapy; HR Hazard ratio; CI Confidential interval; ASA American Society of Anesthesiologists; CEA Carcinoembryonic Antigen; CA19–9 Carbohydrate Antigen 19–9

Predictive nomogram for pCR and decision curve analysis

By incorporating the significant determinants in the Logistic regression analysis, we developed a predictive nomogram for pCR in LARC patients after NCRT as shown in Fig. 2a. The c-index of the nomogram was 0.70 (95% CI 0.70–0.71). The calibration curve (Fig. 2b) presented good statistical performance upon internal validation between predicted and actual pCR rates. DCA was used to evaluate the performance of the nomogram. As shown in Fig. 2c, the model including age group provided more predictive power than either the pCR scheme or the non-pCR scheme. The clinical impact curve (Fig. 2d) shows the prediction of risk stratification of 1000 patients using a resampling bootstrap method.

The association of CD133 expression with survival, pCR rate, and young age

We further investigated the reason for the lower pCR rates in young LARC patients by examining CSC frequency (based on CD133 expression, Fig. 3e, f, g and h). A total of 169 LARC patients were eligible for immunohistochemical analysis. Since the CD133 expression scores were continuous variables, the X-tile program was utilized to identify the optimal cut-off points for determining the greatest actuarial survival difference. Using this method 11 was identified as the cut-off value for CD133 expression (Fig. 3a and b). Based on these cut-off points for CD133 expression, we divided the cohort into low (n = 127) and high (n = 42) subgroups in terms of OS and DFS.
Higher CD133 scores were associated with poorer prognosis in LARC patients following NCRT. The 3-year OS rate for the low CD133 group was significantly higher than that in the high CD133 group (95.1% vs. 62.9%; P < 0.01, Fig. 3d). Moreover, lower CD133 scores were correlated with improved DFS (Fig. 3c). The 3-year DFS rate for the low CD133 group was significantly higher than that in the high CD133 group (92.8% vs. 50.7%; P < 0.01). As shown in Fig. 3i, young patients had a higher CD133 expression score compared to that of old patients (P < 0.01). Moreover, correlation analysis demonstrated that CD133 expression was associated with patient age (P < 0.01, Fig. 3j). Moreover, we examined the CD133 expression in the pCR and non-pCR groups, the results demonstrated that the CD133 expression value in the pCR group was significantly lower than in the non-pCR group (P < 0.01, Fig. 3k). Taken together, these findings indicated that young LARC patients were associated with a higher CD133+ CSC burden, which might contribute to the lower pCR rates.

Discussion

Age is an important factor affecting the efficacy of NCRT in rectal cancer patients. Few studies have focused on young LARC patients following NCRT. In the present study, we explored the efficacy of NCRT in young (< 40 years) and old (≥40 years) LARC patients. The results demonstrated a lower pCR rate in young LARC patients compared with that in old patients, without affecting postoperative complications. For the first time, our study demonstrated that young patients have a higher proportion of CSCs (CD133+), which might contribute to the lower pCR rates.
Young LARC patients often present with aggressive pathological features and advanced stage compared with older patients [46]. Additionally, aggressive pathological features could result in a poorer response to NCRT [2228]. Our results are consistent with those reported by Li et.al [29], in which analysis of the Surveillance, Epidemiology, and End Results (SEER) population-based database revealed that young patients had a better prognosis than old patients. However, the prognosis was inconsistent with the pathological results in their study, because young patients had more aggressive pathological features compared with old patients. This discrepancy may be explained that by the lack of cancer therapy information, including neoadjuvant and adjuvant treatment, and quality of surgery, in the SEER database, all of which are factors that play an important role in patient survival outcome. When treating young LARC patients with rectal cancer, it is important to determine how aggressive the tumor is, so that patients can be informed about the advantages and disadvantages of the treatment. In our study, we demonstrated that young LARC patients displayed poorer pathological features, such as a higher probability of mucinous or signet ring cell and poorly differentiated tumors, which is in accordance with the findings of previous studies [23, 24, 26]. These results indicate that young LARC patients have more severe pathological features than those of older patients.
Age is an important factor for survival benefits and the risk of complications in cancer patients following CRT. To clarify the influence of age on tumor response to NCRT in LARC patients, we performed a logistic regression analysis. pCR has been proposed as a surrogate endpoint for the efficacy of NCRT and oncological outcome in LARC patients. Our results demonstrated that young age is an independent predictive factor for pCR in LARC patients, as well as tumor size, pre-NCRT clinical N stage, and post-NCRT CEA level. Our results also indicated that young LARC patients have poor responses to NCRT, in terms of lower pCR rates. Therefore, more chemotherapy treatment options could be considered in the NCRT regimens in young LARC patients to increase the pCR rate. We further developed a nomogram for predicting pCR to facilitate the decision-making regarding organ-preserving strategies. Our results showed that decisions based on the pCR predictive nomogram yielded more favorable clinical consequences than the treat-all patient and treat-none schemes, even given an extremely small probability threshold. Additionally, the nomogram including age group had superior predictive capability than the nomogram excluding age group.
CSCs are the tumor-initiating cells that are responsible for tumorigenesis [30, 31]. Accumulating evidence has demonstrated that CSCs contribute to resistance to either chemotherapy or radiotherapy in various cancers, including rectal cancer [3234]. It has been reported that cells expressing CD133, which is a putative marker of CSCs, are more resistant to radiochemotherapy than CD133- tumor cells in rectal cancer [32, 33]. Thus, CD133 expression in the tumor cells was detected as a CSC biomarker in LARC patients. However, the association between CSCs and treatment response in young LARC patients remains unclear. Herein, we hypothesized that young LARC patients have more CSCs, resulting in the resistance to NCRT. In this study, we demonstrated higher CD133 expression in cancer tissue of young LARC patients before NCRT compared with that in the old group. Additionally, higher CD133 expression was correlated with a worse prognosis. Together, these findings suggest that young LARC patients have a higher CD133+ CSC burden, contributing to the lower pCR rates. Nevertheless, prospective studies with large sample sizes are required to confirm the role of CD133 in resistance to NCRT.
Several limitations of our study should be noted. First, our retrospective study was subject to potential selection bias. Second, age-related comorbidities, such as the Charlson comorbidity index, were not evaluated due to the lack of adequate data. Third, the impact of gene profiling on response to NCRT was not assessed owing to the lack of complete medical records in some cases. Fourth, CD133 expression was assessed only in some patients owing to the lack of adequate pretreatment specimens. Nevertheless, our study adds to our understanding of the impact of young age on the efficacy of NCRT in patients with LARC.

Conclusions

In this cohort study of 901 LARC patients treated at a single high-volume cancer center, young age (< 40 years) was identified as a significant determinant for predicting pCR in LARC patients following NCRT. Moreover, for the first time, we have demonstrated that young patients have a higher proportion of CSCs (CD133+) than older patients. Larger-scale prospective studies are warranted to confirm our findings.

Acknowledgments

The authors thank all the staff in Department of colorectal surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, People’s Republic of China.
All had patients provided written informed consent for the scientific use of the clinical tissue samples informed consent for inclusion before they participated in the study. The study was conducted in accordance with the Declaration of Helsinki, and the protocol was approved by the Ethics Committee of Fujian Medical University Union Hospital.
Not applicable.

Competing interests

The authors declare that they have no conflict of interest.
Open AccessThis 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/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67:7–30. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67:7–30.
2.
Zurück zum Zitat Cai X, Wu H, Peng J, Zhu J, Cai S, Cai G, Zhang Z. Tolerability and outcomes of radiotherapy or chemoradiotherapy for rectal cancer in elderly patients aged 70 years and older. Radiat Oncol. 2013;8:86.CrossRef Cai X, Wu H, Peng J, Zhu J, Cai S, Cai G, Zhang Z. Tolerability and outcomes of radiotherapy or chemoradiotherapy for rectal cancer in elderly patients aged 70 years and older. Radiat Oncol. 2013;8:86.CrossRef
3.
Zurück zum Zitat Rutten H, den Dulk M, Lemmens V, Nieuwenhuijzen G, Krijnen P, Jansen-Landheer M, van de Poll FL, Coebergh JW, Martijn H, Marijnen C, van de Velde C. Survival of elderly rectal cancer patients not improved: analysis of population based data on the impact of TME surgery. Eur J Cancer. 2007;43:2295–300.CrossRef Rutten H, den Dulk M, Lemmens V, Nieuwenhuijzen G, Krijnen P, Jansen-Landheer M, van de Poll FL, Coebergh JW, Martijn H, Marijnen C, van de Velde C. Survival of elderly rectal cancer patients not improved: analysis of population based data on the impact of TME surgery. Eur J Cancer. 2007;43:2295–300.CrossRef
4.
Zurück zum Zitat Patel SG, Ahnen DJ. Colorectal Cancer in the young. Curr Gastroenterol Rep. 2018;20:15.CrossRef Patel SG, Ahnen DJ. Colorectal Cancer in the young. Curr Gastroenterol Rep. 2018;20:15.CrossRef
5.
Zurück zum Zitat O'Connell JB, Maggard MA, Liu JH, Etzioni DA, Ko CY. Are survival rates different for young and older patients with rectal cancer. Dis Colon Rectum. 2004;47:2064–9.CrossRef O'Connell JB, Maggard MA, Liu JH, Etzioni DA, Ko CY. Are survival rates different for young and older patients with rectal cancer. Dis Colon Rectum. 2004;47:2064–9.CrossRef
6.
Zurück zum Zitat Gallagher EG, Zeigler MG. Rectal carcinoma in patients in the second and third decades of life. Am J Surg. 1972;124:655–9.CrossRef Gallagher EG, Zeigler MG. Rectal carcinoma in patients in the second and third decades of life. Am J Surg. 1972;124:655–9.CrossRef
7.
Zurück zum Zitat Roh MS, Colangelo LH, O'Connell MJ, Yothers G, Deutsch M, Allegra CJ, Kahlenberg MS, Baez-Diaz L, Ursiny CS, Petrelli NJ, Wolmark N. Preoperative multimodality therapy improves disease-free survival in patients with carcinoma of the rectum: NSABP R-03. J Clin Oncol. 2009;27:5124–30.CrossRef Roh MS, Colangelo LH, O'Connell MJ, Yothers G, Deutsch M, Allegra CJ, Kahlenberg MS, Baez-Diaz L, Ursiny CS, Petrelli NJ, Wolmark N. Preoperative multimodality therapy improves disease-free survival in patients with carcinoma of the rectum: NSABP R-03. J Clin Oncol. 2009;27:5124–30.CrossRef
8.
Zurück zum Zitat Sauer R, Liersch T, Merkel S, Fietkau R, Hohenberger W, Hess C, Becker H, Raab HR, Villanueva MT, Witzigmann H, Wittekind C, Beissbarth T, Rödel C. Preoperative versus postoperative chemoradiotherapy for locally advanced rectal cancer: results of the German CAO/ARO/AIO-94 randomized phase III trial after a median follow-up of 11 years. J Clin Oncol. 2012;30:1926–33.CrossRef Sauer R, Liersch T, Merkel S, Fietkau R, Hohenberger W, Hess C, Becker H, Raab HR, Villanueva MT, Witzigmann H, Wittekind C, Beissbarth T, Rödel C. Preoperative versus postoperative chemoradiotherapy for locally advanced rectal cancer: results of the German CAO/ARO/AIO-94 randomized phase III trial after a median follow-up of 11 years. J Clin Oncol. 2012;30:1926–33.CrossRef
9.
Zurück zum Zitat van Gijn W, Marijnen CA, Nagtegaal ID, Kranenbarg EM, Putter H, Wiggers T, Rutten HJ, Påhlman L, Glimelius B, van de Velde CJ. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer: 12-year follow-up of the multicentre, randomised controlled TME trial. Lancet Oncol. 2011;12:575–82.CrossRef van Gijn W, Marijnen CA, Nagtegaal ID, Kranenbarg EM, Putter H, Wiggers T, Rutten HJ, Påhlman L, Glimelius B, van de Velde CJ. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer: 12-year follow-up of the multicentre, randomised controlled TME trial. Lancet Oncol. 2011;12:575–82.CrossRef
10.
Zurück zum Zitat Park IJ, You YN, Agarwal A, Skibber JM, Rodriguez-Bigas MA, Eng C, Feig BW, Das P, Krishnan S, Crane CH, Hu CY, Chang GJ. Neoadjuvant treatment response as an early response indicator for patients with rectal cancer. J Clin Oncol. 2012;30:1770–6.CrossRef Park IJ, You YN, Agarwal A, Skibber JM, Rodriguez-Bigas MA, Eng C, Feig BW, Das P, Krishnan S, Crane CH, Hu CY, Chang GJ. Neoadjuvant treatment response as an early response indicator for patients with rectal cancer. J Clin Oncol. 2012;30:1770–6.CrossRef
11.
Zurück zum Zitat Maas M, Nelemans PJ, Valentini V, Das P, Rödel C, Kuo LJ, Calvo FA, García-Aguilar J, Glynne-Jones R, Haustermans K, Mohiuddin M, Pucciarelli S, Small W, Suárez J, Theodoropoulos G, Biondo S, Beets-Tan RG, Beets GL. Long-term outcome in patients with a pathological complete response after chemoradiation for rectal cancer: a pooled analysis of individual patient data. Lancet Oncol. 2010;11:835–44.CrossRef Maas M, Nelemans PJ, Valentini V, Das P, Rödel C, Kuo LJ, Calvo FA, García-Aguilar J, Glynne-Jones R, Haustermans K, Mohiuddin M, Pucciarelli S, Small W, Suárez J, Theodoropoulos G, Biondo S, Beets-Tan RG, Beets GL. Long-term outcome in patients with a pathological complete response after chemoradiation for rectal cancer: a pooled analysis of individual patient data. Lancet Oncol. 2010;11:835–44.CrossRef
12.
Zurück zum Zitat Neugut AI, Fleischauer AT, Sundararajan V, Mitra N, Heitjan DF, Jacobson JS, Grann VR. Use of adjuvant chemotherapy and radiation therapy for rectal cancer among the elderly: a population-based study. J Clin Oncol. 2002;20:2643–50.CrossRef Neugut AI, Fleischauer AT, Sundararajan V, Mitra N, Heitjan DF, Jacobson JS, Grann VR. Use of adjuvant chemotherapy and radiation therapy for rectal cancer among the elderly: a population-based study. J Clin Oncol. 2002;20:2643–50.CrossRef
13.
Zurück zum Zitat Cafiero F, Gipponi M, Lionetto R. Randomised clinical trial of adjuvant postoperative RT vs. sequential postoperative RT plus 5-FU and levamisole in patients with stage II-III resectable rectal cancer: a final report. J Surg Oncol. 2003;83:140–6.CrossRef Cafiero F, Gipponi M, Lionetto R. Randomised clinical trial of adjuvant postoperative RT vs. sequential postoperative RT plus 5-FU and levamisole in patients with stage II-III resectable rectal cancer: a final report. J Surg Oncol. 2003;83:140–6.CrossRef
14.
Zurück zum Zitat Vermeulen L. de Sousa e Melo F, Richel DJ, Medema JP. The developing cancer stem-cell model: clinical challenges and opportunities. Lancet Oncol. 2012;13:e83–9.CrossRef Vermeulen L. de Sousa e Melo F, Richel DJ, Medema JP. The developing cancer stem-cell model: clinical challenges and opportunities. Lancet Oncol. 2012;13:e83–9.CrossRef
15.
Zurück zum Zitat Boman BM, Wicha MS. Cancer stem cells: a step toward the cure. J Clin Oncol. 2008;26:2795–9.CrossRef Boman BM, Wicha MS. Cancer stem cells: a step toward the cure. J Clin Oncol. 2008;26:2795–9.CrossRef
16.
Zurück zum Zitat Dick JE. Looking ahead in cancer stem cell research. Nat Biotechnol. 2009;27:44–6.CrossRef Dick JE. Looking ahead in cancer stem cell research. Nat Biotechnol. 2009;27:44–6.CrossRef
17.
Zurück zum Zitat Ryan R, Gibbons D, Hyland JM, Treanor D, White A, Mulcahy HE, O'Donoghue DP, Moriarty M, Fennelly D, Sheahan K. Pathological response following long-course neoadjuvant chemoradiotherapy for locally advanced rectal cancer. Histopathology. 2005;47:141–6.CrossRef Ryan R, Gibbons D, Hyland JM, Treanor D, White A, Mulcahy HE, O'Donoghue DP, Moriarty M, Fennelly D, Sheahan K. Pathological response following long-course neoadjuvant chemoradiotherapy for locally advanced rectal cancer. Histopathology. 2005;47:141–6.CrossRef
18.
Zurück zum Zitat Zhang Y, Xu Z, Sun Y, Chi P, Lu X. Knockdown of KLK11 reverses oxaliplatin resistance by inhibiting proliferation and activating apoptosis via suppressing the PI3K/AKT signal pathway in colorectal cancer cell. Onco Targets Ther. 2018;11:809–21.CrossRef Zhang Y, Xu Z, Sun Y, Chi P, Lu X. Knockdown of KLK11 reverses oxaliplatin resistance by inhibiting proliferation and activating apoptosis via suppressing the PI3K/AKT signal pathway in colorectal cancer cell. Onco Targets Ther. 2018;11:809–21.CrossRef
19.
Zurück zum Zitat Rousson V, Zumbrunn T. Decision curve analysis revisited: overall net benefit, relationships to ROC curve analysis, and application to case-control studies. BMC Med Inform Decis Mak. 2011;11:45.CrossRef Rousson V, Zumbrunn T. Decision curve analysis revisited: overall net benefit, relationships to ROC curve analysis, and application to case-control studies. BMC Med Inform Decis Mak. 2011;11:45.CrossRef
20.
Zurück zum Zitat Vickers AJ, Elkin EB. Decision curve analysis: a novel method for evaluating prediction models. Med Decis Mak. 2006;26:565–74.CrossRef Vickers AJ, Elkin EB. Decision curve analysis: a novel method for evaluating prediction models. Med Decis Mak. 2006;26:565–74.CrossRef
21.
Zurück zum Zitat Camp RL, Dolled-Filhart M, Rimm DL. X-tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res. 2004;10:7252–9.CrossRef Camp RL, Dolled-Filhart M, Rimm DL. X-tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res. 2004;10:7252–9.CrossRef
22.
Zurück zum Zitat Barbaro B, Leccisotti L, Vecchio FM, Di MM, Serra T, Salsano M, Poscia A, Coco C, Persiani R, Alfieri S, Gambacorta MA, Valentini V, Giordano A, Bonomo L. The potential predictive value of MRI and PET-CT in mucinous and nonmucinous rectal cancer to identify patients at high risk of metastatic disease. Br J Radiol. 2017;90:20150836.CrossRef Barbaro B, Leccisotti L, Vecchio FM, Di MM, Serra T, Salsano M, Poscia A, Coco C, Persiani R, Alfieri S, Gambacorta MA, Valentini V, Giordano A, Bonomo L. The potential predictive value of MRI and PET-CT in mucinous and nonmucinous rectal cancer to identify patients at high risk of metastatic disease. Br J Radiol. 2017;90:20150836.CrossRef
23.
Zurück zum Zitat Reggiani BL, Lionti S, Domati F, Pagliani G, Mattioli E, Barresi V. Histological grading based on poorly differentiated clusters is predictive of tumour response and clinical outcome in rectal carcinoma treated with neoadjuvant chemoradiotherapy. Histopathology. 2017;71:393–405.CrossRef Reggiani BL, Lionti S, Domati F, Pagliani G, Mattioli E, Barresi V. Histological grading based on poorly differentiated clusters is predictive of tumour response and clinical outcome in rectal carcinoma treated with neoadjuvant chemoradiotherapy. Histopathology. 2017;71:393–405.CrossRef
24.
Zurück zum Zitat Sengul N, Wexner SD, Woodhouse S, Arrigain S, Xu M, Larach JA, Ahn BK, Weiss EG, Nogueras JJ, Berho M. Effects of radiotherapy on different histopathological types of rectal carcinoma. Color Dis. 2006;8:283–8.CrossRef Sengul N, Wexner SD, Woodhouse S, Arrigain S, Xu M, Larach JA, Ahn BK, Weiss EG, Nogueras JJ, Berho M. Effects of radiotherapy on different histopathological types of rectal carcinoma. Color Dis. 2006;8:283–8.CrossRef
25.
Zurück zum Zitat Grillo-Ruggieri F, Mantello G, Berardi R, Cardinali M, Fenu F, Iovini G, Montisci M, Fabbietti L, Marmorale C, Guerrieri M, Saba V, Bearzi I, Mattioli R, Bonsignori M, Cascinu S. Mucinous rectal adenocarcinoma can be associated to tumor downstaging after preoperative chemoradiotherapy. Dis Colon Rectum. 2007;50:1594–603.CrossRef Grillo-Ruggieri F, Mantello G, Berardi R, Cardinali M, Fenu F, Iovini G, Montisci M, Fabbietti L, Marmorale C, Guerrieri M, Saba V, Bearzi I, Mattioli R, Bonsignori M, Cascinu S. Mucinous rectal adenocarcinoma can be associated to tumor downstaging after preoperative chemoradiotherapy. Dis Colon Rectum. 2007;50:1594–603.CrossRef
26.
Zurück zum Zitat Shin US, Yu CS, Kim JH, Kim TW, Lim SB, Yoon SN, Yoon YS, Kim CW, Kim JC. Mucinous rectal cancer: effectiveness of preoperative chemoradiotherapy and prognosis. Ann Surg Oncol. 2011;18:2232–9.CrossRef Shin US, Yu CS, Kim JH, Kim TW, Lim SB, Yoon SN, Yoon YS, Kim CW, Kim JC. Mucinous rectal cancer: effectiveness of preoperative chemoradiotherapy and prognosis. Ann Surg Oncol. 2011;18:2232–9.CrossRef
27.
Zurück zum Zitat Yu SK, Chand M, Tait DM, Brown G. Magnetic resonance imaging defined mucinous rectal carcinoma is an independent imaging biomarker for poor prognosis and poor response to preoperative chemoradiotherapy. Eur J Cancer. 2014;50:920–7.CrossRef Yu SK, Chand M, Tait DM, Brown G. Magnetic resonance imaging defined mucinous rectal carcinoma is an independent imaging biomarker for poor prognosis and poor response to preoperative chemoradiotherapy. Eur J Cancer. 2014;50:920–7.CrossRef
28.
Zurück zum Zitat Oberholzer K, Menig M, Kreft A, Schneider A, Junginger T, Heintz A, Kreitner KF, Hötker AM, Hansen T, Düber C, Schmidberger H. Rectal cancer: mucinous carcinoma on magnetic resonance imaging indicates poor response to neoadjuvant chemoradiation. Int J Radiat Oncol Biol Phys. 2012;82:842–8.CrossRef Oberholzer K, Menig M, Kreft A, Schneider A, Junginger T, Heintz A, Kreitner KF, Hötker AM, Hansen T, Düber C, Schmidberger H. Rectal cancer: mucinous carcinoma on magnetic resonance imaging indicates poor response to neoadjuvant chemoradiation. Int J Radiat Oncol Biol Phys. 2012;82:842–8.CrossRef
29.
Zurück zum Zitat Li Q, Cai G, Li D, Wang Y, Zhuo C, Cai S. Better long-term survival in young patients with non-metastatic colorectal cancer after surgery, an analysis of 69,835 patients in SEER database. PLoS One. 2014;9:e93756.CrossRef Li Q, Cai G, Li D, Wang Y, Zhuo C, Cai S. Better long-term survival in young patients with non-metastatic colorectal cancer after surgery, an analysis of 69,835 patients in SEER database. PLoS One. 2014;9:e93756.CrossRef
30.
Zurück zum Zitat Papaccio F, Paino F, Regad T, Papaccio G, Desiderio V, Tirino V. Concise review: Cancer cells, Cancer stem cells, and Mesenchymal stem cells: influence in Cancer development. Stem Cells Transl Med. 2017;6:2115–25.CrossRef Papaccio F, Paino F, Regad T, Papaccio G, Desiderio V, Tirino V. Concise review: Cancer cells, Cancer stem cells, and Mesenchymal stem cells: influence in Cancer development. Stem Cells Transl Med. 2017;6:2115–25.CrossRef
31.
Zurück zum Zitat Ramos EK, Hoffmann AD, Gerson SL, Liu H. New opportunities and challenges to defeat Cancer stem cells. Trends Cancer. 2017;3:780–96.CrossRef Ramos EK, Hoffmann AD, Gerson SL, Liu H. New opportunities and challenges to defeat Cancer stem cells. Trends Cancer. 2017;3:780–96.CrossRef
32.
Zurück zum Zitat Sprenger T, Conradi LC, Beissbarth T, Ermert H, Homayounfar K, Middel P, Rüschoff J, Wolff HA, Schüler P, Ghadimi BM, Rödel C, Becker H, Rödel F, Liersch T. Enrichment of CD133-expressing cells in rectal cancers treated with preoperative radiochemotherapy is an independent marker for metastasis and survival. Cancer. 2013;119:26–35.CrossRef Sprenger T, Conradi LC, Beissbarth T, Ermert H, Homayounfar K, Middel P, Rüschoff J, Wolff HA, Schüler P, Ghadimi BM, Rödel C, Becker H, Rödel F, Liersch T. Enrichment of CD133-expressing cells in rectal cancers treated with preoperative radiochemotherapy is an independent marker for metastasis and survival. Cancer. 2013;119:26–35.CrossRef
33.
Zurück zum Zitat Saigusa S, Tanaka K, Toiyama Y, Yokoe T, Okugawa Y, Ioue Y, Miki C, Kusunoki M. Correlation of CD133, OCT4, and SOX2 in rectal cancer and their association with distant recurrence after chemoradiotherapy. Ann Surg Oncol. 2009;16:3488–98.CrossRef Saigusa S, Tanaka K, Toiyama Y, Yokoe T, Okugawa Y, Ioue Y, Miki C, Kusunoki M. Correlation of CD133, OCT4, and SOX2 in rectal cancer and their association with distant recurrence after chemoradiotherapy. Ann Surg Oncol. 2009;16:3488–98.CrossRef
34.
Zurück zum Zitat Fan CW, Chen T, Shang YN, Gu YZ, Zhang SL, Lu R, OuYang SR, Zhou X, Li Y, Meng WT, Hu JK, Lu Y, Sun XF, Bu H, Zhou ZG, Mo XM. Cancer-initiating cells derived from human rectal adenocarcinoma tissues carry mesenchymal phenotypes and resist drug therapies. Cell Death Dis. 2013;4:e828.CrossRef Fan CW, Chen T, Shang YN, Gu YZ, Zhang SL, Lu R, OuYang SR, Zhou X, Li Y, Meng WT, Hu JK, Lu Y, Sun XF, Bu H, Zhou ZG, Mo XM. Cancer-initiating cells derived from human rectal adenocarcinoma tissues carry mesenchymal phenotypes and resist drug therapies. Cell Death Dis. 2013;4:e828.CrossRef
Metadaten
Titel
Worse treatment response to neoadjuvant chemoradiotherapy in young patients with locally advanced rectal cancer
verfasst von
Yiyi Zhang
Liangliang Yan
Yong Wu
Meifang Xu
Xing Liu
Guoxian Guan
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2020
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-020-07359-2

Weitere Artikel der Ausgabe 1/2020

BMC Cancer 1/2020 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Update Onkologie

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