Skip to main content
Erschienen in: Annals of Surgical Oncology 4/2020

Open Access 29.10.2019 | Gastrointestinal Oncology

Prophylactic HIPEC in pT4 Colon Tumors: Proactive Approach or Overtreatment?

verfasst von: Nerea Borda Arrizabalaga, MD, José María Enriquez Navascués, PhD, Garazi Elorza Echaniz, MD, Yolanda Saralegui Ansorena, PhD, Carlos Placer Galán, PhD, Xabier Arteaga Martín, PhD, Leyre Velaz Pardo, MD

Erschienen in: Annals of Surgical Oncology | Ausgabe 4/2020

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

The peritoneum is the second most common site for metastasis in patients with colorectal cancer. Various factors have been studied to identify patients at risk of developing peritoneal carcinomatosis (PC), including T4 tumors. The objectives were to assess the incidence of synchronous and metachronous PC, explore potential risk factors for developing PC as the only site of metastasis, and identify which patients might be candidates for prophylactic hyperthermic intraperitoneal chemotherapy (HIPEC).

Methods

We conducted a retrospective analysis of 125 patients with pT4 colon cancer who underwent surgery in a single center between January 2010 and December 2014.

Results

Of the 947 colon cancer patients who underwent surgery, 125 (13.2%) were diagnosed with pT4a or b colon carcinoma. The median follow-up was 3.7 years. The overall rate of PC was 34.3%, being synchronous in 12% and metachronous in 22.3% of cases. The 8% and 6% of synchronous and metachronous cases of PC respectively were isolated (single site) metastasis. The incidence of PC was 6.1% at 1 year and 14.5% at 3 years after surgery. pT4 was not found to be an independent risk factor for the development of PC (p = 0.231). Nonetheless, the rate of metachronous PC as a single site of metastasis was higher in patients with pT4 tumors and peritoneal nodules around the primary tumor and/or tumor perforation (p = 0.027) and/or who underwent emergency surgery (p = 0.043) than other patients.

Conclusions

Considering pT4 tumor stage as the only risk factor for the development of PC in deciding whether to administer prophylactic HIPEC would lead to unjustified overtreatment.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Worldwide, colorectal cancer is the third most common cause of cancer in men and the second in women. It is expected to affect some 2.4 million individuals by 2034.1 The peritoneum is the second most common site of metastasis in patients with colorectal cancer, accounting for 25–35% of all cases of recurrence.2,3 Among patients with recurrent disease, 5–10% have synchronous and 20–50% develop metachronous peritoneal carcinomatosis (PC).47
The treatment for PC has evolved greatly over the past 15 years. The goal of the treatment has changed from being purely palliative or supportive to being considered curative in selected patients. The combination of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) has achieved median survival rates of up to 64 months; peritoneal cancer index (PCI) and complete surgical cytoreduction (CRS-0) are the two main predictors of prognosis with this strategy.815 Nonetheless, CRS and HIPEC are not risk-free, with nonnegligible associated morbidity and mortality.16,17
Numerous clinical, pathological, and biological factors have been studied to identify patients with the highest risk of having synchronous or developing metachronous PC and to be able to diagnose it early, while the PCI is still low, and have an impact on outcomes in this population.1820 With the same goal of altering the natural history of PC, two different strategies have been proposed: one consisting of administering prophylactic HIPEC at the time of primary tumor surgery to prevent peritoneal recurrence, and the other performing systematic second-look surgery with HIPEC, preempting peritoneal recurrence.18
In addition to spontaneous or iatrogenic tumor perforation, peritoneal tumors around the primary tumor and ovarian metastases, the presence of tumors at stage T4 is a potential risk factor for the development of PC.18 It has been estimated that up to 26% of patients with T4 tumors would benefit from either of the aforementioned strategies.21 Nonetheless, the real incidence of PC as an isolated finding in stage T4 disease is debated and we may be overestimating the problem. On the one hand, preoperatively, based on imaging alone, T4 tumors may be overdiagnosed, and on the other, if the incidence of metachromous PC were lower than expected use of the prophylactic strategies would result in overtreatment, with the associated morbidity.
The primary objective of this study was to assess the incidence of synchronous and metachronous PC in pT4 colon cancer patients who underwent surgery and were followed-up in an integrated healthcare organization, with medical records and responsibility for patient monitoring shared between hospital and primary care providers. The secondary objectives were to explore potential risk factors for the development of PC as the only site of metastasis and identify which patients might be candidates for prophylactic HIPEC.

Methods

We performed a retrospective analysis of pT4 colon cancer patients, regardless of whether they had lymph node involvement and/or distant metastasis, who underwent surgery between January 2010 and December 2014 in a healthcare region with integrated social and primary and specialist health care, with a shared follow-up protocol, and with no missing data. The study protocol was approved by the scientific research ethics committee of the institution.
The TNM cancer staging was based on the Seventh Edition of the American Joint Committee on Cancer Colon and Rectum Cancer Staging Manual. Accordingly, T4a tumors were defined as those with serosal involvement and T4b tumors as those in which the tumor has spread to neighboring organs.
We recorded data on epidemiological, clinical, surgical, and histopathological characteristics, the initial staging and findings during follow-up. The metachronous PC was diagnosed by imaging tests: abdominal ultrasound and CT scans. These tests were performed every 6 months (at least one CT scan per year) for the first 3 years and annually until the 5-year mark. Criteria for establishing metachronous PC included: histologic or cytologic confirmation, palpable disease, disease evident on radiographic studies with subsequent image or clinical progression, and supportive biochemical data, i.e., rising level of carcinoembryonic antigen (CEA). No patient was accepted as having PC by virtue of one interval CT, alone. A colonoscopy was performed at 3 years after the surgical intervention (or at 1 year if no previous colonoscopy had been complete).

Statistical Analysis

Qualitative data were expressed as percentages and absolute values and compared with the χ2 test. In the case of quantitative variables, first, the Kolmogorov–Smirnov test was used to characterize the distribution, and then data were expressed as means and standard deviations and compared using Student’s t test if normally distributed, and otherwise, they were expressed as medians and interquartile ranges and compared using the Mann–Whitney U test.
Data on overall survival, disease-free survival and time to progression were analyzed using Kaplan–Meier curves, and factors were compared using log-rank tests. The variables with a significance level ≤ 0.2 were included in the binary logistic regression to explore which were associated with prognosis including COX-2.
Survival was measured from the date of the surgical intervention for the primary tumor until the date of the last follow-up or until the patient died. p values < 0.05 were considered statistically significant.

Results

A total of 947 patients underwent colon cancer surgery at our institution between January 2010 and December 2014. Of these, 125 patients (13.2%) were diagnosed with pT4a or b colon carcinoma. Data were collected until 31 December 2017, with a median follow-up of 3.7 (range 1–7.7) years.
The main clinical characteristics of the patients and histopathological characteristics of the tumors are described in Table 1. As can be observed, a quarter (25.6%) of patients had PC, distant metastasis, or both at the time of diagnosis. Overall, 12% had synchronous PC, 8% corresponding to patients with isolated dissemination, and 4% to patients who also had distant metastasis. Having a primary tumor that has spread to neighboring organs (pT4b) and lymph node involvement were identified as risk factors for synchronous PC (Table 2).
Table 1
Clinical characteristics of patients and histopathological characteristics of the tumors
Mean age
67 years old (32–91)
Sex
 Male
71 (56.8)
 Female
54 (43.2)
Site of primary tumora
 Left colon
62 (49.6)
 Transverse colon
10 (8.0)
 Right colon
53 (42.4)
Type of surgery
 Scheduled
96 (76.8)
 Emergency
29 (23.2)
Spontaneous/Iatrogenic perforation of the tumor
 No
88 (70.4)
 Yes
37 (29.6)
Obstruction of the primary tumor
 No
98 (78.4)
 Yes
27 (21.6)
Type of resection
 R0
110 (88)
 R1
4 (3.2)
 R2
11 (8.8)
Degree of tumor differentiation
 G1
36 (28.8)
 G2
70 (56.0)
 G3
19 (15.2)
Histological findings
 Adenocarcinoma
110 (88.0)
 Mucinous adenocarcinoma
10 (8.0)
 Signet ring cell carcinoma
5 (4.0)
pT4
 pT4a
81 (64.8)
 pT4b
44 (35.2)
pN
 pN0
52 (41.6)
 pN1
44 (35.2)
  pN1a
19 (15.2)
  pN1b
24 (19.2)
  pN1c
1 (0.8)
 pN2
29 (23.2)
  pN2a
17 (13.6)
  pN2b
12 (9.6)
M
 M0
93 (74.4)
 M1
32 (25.6)
Tumor stage
 IIa
28 (22.4)
 IIb
19 (15.2)
 IIIb
24 (19.2)
 IIIc
23 (18.4)
 IVa
23 (18.4)
 IVb
8 (6.4)
Anastomotic leakage
 No
113 (90.4)
 Yes
12 (9.6)
Site of metastasis at primary tumor diagnosis
32 (25.6)
 Visceral metastases
17 (13.6)
 Peritoneal carcinomatosis
10 (8.0)
 Visceral metastases + peritoneal carcinomatosis
5 (4.0)
aLeft colon: sigmoid colon, descending colon, splenic flexure; right colon: caecum, ascending colon, hepatic flexure
Table 2
Risk factors for synchronous peritoneal carcinomatosis (univariate analysis)
 
Peritoneal carcinomatosis (%)
p
Site of the primary tumora
0.482
 Left colon
6/62 (9.7)
 
 Transverse colon
0/9 (0.0)
 
 Right colon
7/54 (12.3)
 
Degree of tumor differentiation
0.255
 G1
3/36 (8.3)
 
 G2
6/70 (8.7)
 
 G3
4/19 (21.1)
 
Transmural involvement (pT)
0.028
 Serosal involvement
12/81(14.8)
 
 Spread to neighboring organs
1/44(2.3)
 
pN
0.026
 pN0
2/52 (3.8)
 
 pN1
3/19 (15.8)
 
  pN1a
3/24 (12.5)
 
  pN1b
1/1 (100)
 
  pN1c
2/17 (11.8)
 
 pN2
1/12 (8.3)
 
  pN2a
  
  pN2b
  
Histological findings
0.473
 Adenocarcinoma
12/116 (10.3)
 
 Mucinous adenocarcinoma
0/5 (0.0)
 
 Signet ring cell carcinoma
1/4 (25.0)
 
aLeft colon: sigmoid colon, descending colon, splenic flexure; right colon: caecum, ascending colon, hepatic flexure
Bold values are statistically significant (p < 0.05)
Table 3 summarizes the course of patients during the follow-up period. A total of 21 patients (22.3%) were found to have PC during follow-up, but only 6 (6.12%) had isolated PC, as a single site of recurrence, being the rates of PC at 1 and 3 years after surgery 6.1% (95% confidence interval [CI] 87.9–97.6%) and 14.5% (95% CI 76.6–91.2%), respectively. During the follow-up, tumor markers increased in 12 of 21 patients (57.2%), and in patients with metachronous PC as the single site of metastasis, increased CEA was observed in 4 of 6 (67%). Most of the patients were asymptomatic. Only three patients presented colic abdominal pain or lumbar pain. The PC was confirmed by surgery or biopsy in only four patients (19.1%).
Table 3
Follow-up
 
N (%)
Development of distant metastases (no PC)
 No
72 (73.4)
 Yes
26 (27.7)
Site of metastases
 Liver
14 (53.8)
 Lung
9 (34.6)
  ≥ 2 organs
3 (11.5)
PC
 No
77 (78.6)
 Yes
21 (22.3)
PC as a single site of metastasis
6 (6.1)
Local recurrence
 No
77 (78.6)
 Yes
21 (21.4)
Mortality
63 (64.3)
Causes of mortality
 Progression of disease
42 (66.7)
 Post-surgical complications
5 (7.8)
 Comorbidities
16 (25.4)
PC peritoneal carcinomatosis
The potential risk factors for PC are summarized in Table 4. Among the variables analyzed, emergency rather than scheduled surgery (p = 0.019), tumor perforation (p = 0.047), and distant metastasis at diagnosis of the primary tumor (p = 0.047) emerged as independent risk factors for the development of metachronous PC, but they did not remain significant in the multivariate analysis (Table 5).
Table 4
Risk factors for developing metachronous peritoneal carcinomatosis (univariate analysis)
 
Peritoneal carcinomatosis (%)
p
Type of resection
0.713
 R0
19/110 (17.3)
 
 R1
1/4 (25.0)
 
 R2
1/11 (9.1)
 
Degree of tumor differentiation
0.697
 G1
7/36 (19.4)
 
 G2
12/70 (17.1)
 
 G3
2/17 (11.8)
 
Histological finding
0.756
 Adenocarcinoma
18/110 (16.36)
 
 Mucinous adenocarcinoma
2/10 (20.0)
 
 Signet ring cell carcinoma
1/5 (20.0)
 
Transmural involvement (pT)
0.231
 pT4a
16/81 (19.7)
 
 pT4b
5/44 (11.4)
 
Lymph node involvement (pN)
0.235
 pN0
6/52 (11.5)
 
 pN1
4/19 (21.1)
 
  pN1a
4/24 (16.7)
 
  pN1b
1/1 (100)
 
  pN1c
3/17 (17.6)
 
 pN2
3/12(25)
 
  pN2a
  
  pN2b
  
Metastasis (M)
0.047
 M0
12/93 (12.9)
 
 M1
9/32 (28.1)
 
Tumor stage
0.371
 Iia
2/28 (7.1)
 
 Iib
2/19 (10.5)
 
 IIIb
2/24 (8.3)
 
 IIIc
3/23 (13)
 
 Iva
6/23 (26.1)
 
 Ivb
6/8 (75)
 
Type of surgery
0.019
 Scheduled
12/96 (12.5)
 
 Emergency
9/29 (31.0)
 
Spontaneous/iatrogenic tumor perforation
10/37 (27.1)
0.047
Primary tumor obstruction
6/27 (22.2)
0.395
Anastomotic leakage
0/12 (9.4)
0.102
Site of primary tumora
0.827
 Left colon
11/62 (17.7)
 
 Transverse colon
2/10 (20)
 
 Right colon
8/53 (15.1)
 
Peritoneal implants around the primary tumor
5/125 (4.0)
0.157
aLeft colon: sigmoid colon, descending colon, splenic flexure; right colon: caecum, ascending colon, hepatic flexure
Bold values are statistically significant (p < 0.05)
Table 5
Multivariate analysis exploring potential risk factors for the development of metachronous peritoneal carcinomatosis (N = 125)
 
p
95% CI
Emergency surgery
0.052
0.168–1.01
Affectation of neighboring organs
0.992
0.295–3.848
pN
0.948
0.198–11.001
pM
0.109
0.824–6.770
Site of the primary tumor
0.799
0.246–2.942
Primary tumor perforation
0.118
0.168–1.221
Primary tumor obstruction
0.908
0.164–5.001
Peritoneal implants around the primary tumor
0.186
0.007–2.637
Anastomotic leakage
0.999
ns
ns no significance
Nonetheless, in the subgroup analysis, patients with pT4 colon tumors with perforation who underwent emergency surgery were at greater risk of developing PC than other patients (p = 0.043), as were patients with pT4 tumors with perforation and peritoneal nodules around the primary tumor (p = 0.027).
The 3- and 5-year overall survival rates were 60% (95% CI 50.8–67.8%) and 50.3% (95% CI 40.9–59%), respectively, with a median survival of 5.4 years. The mortality rate was 14 deaths/100/year. Five patients (7.8%) died in the postoperative period (2–3 months) due to complications related to the surgical intervention, 42 (66.7%) died due to disease progression, and 16 (25.4%) died due to other causes, namely, patient-related comorbidities.

Discussion

In our study, the overall rate of PC in patients with pT4 colon carcinoma was 34.3%: 12% corresponding to synchronous and 22.3% to metachronous dissemination, respectively. On the other hand, the dissemination was only isolated (a single site) in 8% and 6% of cases of synchronous and metachronous PC, respectively. The incidence rate of PC was 6.1% at 1 year and 14.5% 3 years after surgery.
The overall incidence of PC in our series was higher than that in other studies, which have found rates of PC of 12–18% in patients with pT4 tumors4,22; however, despite complete clinical follow-up, the incidence of PC as a single site of metachronous metastasis was lower (6.1%) than in other research, which has indicated isolated PC in up to a third of patients, with rates of local and peritoneal recurrence of 15.6% and 36.7% at 1 and 3 years after surgery for the primary tumor, respectively.21 Notably, the overall survival in our series is similar to that found previously in consecutive nonselected patients with pT4 colon cancer.21 This leads us to believe that the series analyzed is representative of the tumor stage.
Procedures for diagnosing PC in the initial stages have considerable limitations. This shortage of effective tools for the early diagnosis of PC may result in an under-diagnosis of PC in cases in which systematic laparotomy/laparoscopy is not performed.
In our study, involvement beyond the serosa and involvement of lymph nodes at the time of primary tumor diagnosis have been identified as potential risk factors for synchronous but not metachronous PC. Regarding the development of metachronous PC, our univariate analysis suggests that emergency surgery and primary tumor perforation may be risk factors, although these findings were not confirmed in the multivariate analysis, likely due to the small sample size. Other possible risk factors directly related to the development of PC after curative colorectal surgery include peritoneal implants around the primary tumor, ovarian metastases, the primary tumor being in the right colon, and pT4 and pN2 stage disease.23,24
Although the prophylactic (surgery for the primary tumor + HIPEC) and preemptive (systematic second-look surgery + HIPEC) strategies are different, they have the common objective of improving oncological outcomes in patients at high risk of developing PC. Prophylactic HIPEC has the great appeal of not requiring an additional intervention, with potential further peritoneal and visceral resections,17 but it does have some downsides. On the one hand, it is not available at all times or in all centers offering surgery for colon cancer, and there is a risk of overtreatment, because it is not possible to identify the involvement of structures or differentiate between cT3 and cT4 intraoperatively. On the other hand, it has some ethical limitations, related to the lack of information and prior patient consent (oophorectomy, complications associated with an experimental treatment that is not free of complications, etc.). These logistic and ethical limitations have led to a clinical trial of the prophylactic strategy being withdrawn before completion of recruitment.25
Second-look surgery with HIPEC seems to be more accessible and appropriate for daily clinical practice. There are at least two currently ongoing clinical trials of preemptive strategies that differ not only in the selection of cases in relation to risk factors for the development of PC but also the optimal timing of the intervention. In the French trial (PROPHYLOCHIP), second-look surgery is performed 6–12 months after surgery for the primary tumor.26 In contrast, in the Dutch trial (COLOPEC), the second-look procedure is performed immediately after surgery for the primary tumor (10 days) or after an interval of 5–8 weeks.20 In both studies, there is consensus on performing this type of intervention in patients with isolated PC and various different risk factors for the peritoneal development, such as those mentioned above, but what about pT4 tumors?
Unexpectedly, both studies confirm that criteria for high risk of developing PC are strong but a proactive strategy, including a systematic second-look surgery plus HIPEC failed to improve survival, in comparison to an adequate surveillance. In addition, a recent French study have suggested that the addition of Oxaliplatin-HIPEC on the top of cytoreductive surgery does not influence both overall survival and disease-free survival but suggest that HIPEC with Oxaliplatin may be beneficial for patients with a medium PCI.1115,27 Therefore, at the present time, the role of HIPEC in the treatment of PC is not clear, and long-term results have to be awaited to assess the role of prophylactic/adjuvant HIPEC.
As reported previously, the overall incidence of PC in our study was 34.3%. This could lead us to believe that one-third of patients might benefit from a proactive approach to avoid the development of PC. Nonetheless, in the group with synchronous PC (12%), 4% had distant metastasis, and of the 22.3% of patients with metachronous PC, 15.9% developed distant metastasis in the follow-up. Therefore, we deduced that prophylactic HIPEC might prevent the development of PC in 14.4% of patients. The other patients would also have distant metastasis.
Various randomized clinical trials currently in the recruitment phase are seeking to assess the oncological efficacy of prophylactic HIPEC in cT4 tumors considering this stage of disease as the only risk factor for the development of PC.20,2830 In our study, although 22.3% of patients developed metachronous PC, only 6.1% of patients had metastasis at this site alone, and pT4 as a single risk factor was not shown to be an independent risk factor for the development of PC. Nonetheless, patients with pT4 tumors and peritoneal implants around the primary tumor and/or tumor perforation and/or who underwent emergency surgery had a higher rate of metachronous PC as a single site of metastasis than other patients.

Conclusions

Considering pT4 tumor stage as the only risk factor for the development of PC in deciding whether to administer prophylactic or preemptive HIPEC would lead to unjustified overtreatment. Knowing that the role of HIPEC in the treatment of PC is not clear, we should wait for the results of the ongoing clinical trials before advocating either prophylactic or preemptive strategies for managing tumors in patients with various risk factors for developing PC.

Disclosure

The authors have no commercial interests to declare and did not receive any external financial or material support.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

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.

Bis 30. April 2024 bestellen und im ersten Jahr nur 199 € zahlen!

e.Med Interdisziplinär

Kombi-Abonnement

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.

Literatur
1.
Zurück zum Zitat Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86.CrossRef Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86.CrossRef
2.
Zurück zum Zitat Brodsky JT, Cohen AM. Peritoneal seeding following potentially curative resection of colonic carcinoma: implications for adjuvant therapy. Dis Colon Rectum. 1991;34:723–7.CrossRef Brodsky JT, Cohen AM. Peritoneal seeding following potentially curative resection of colonic carcinoma: implications for adjuvant therapy. Dis Colon Rectum. 1991;34:723–7.CrossRef
3.
Zurück zum Zitat Elferink MAG, de Jong KP, Klaase JM, et al. Metachronous metastases from colorectal cancer: a population-based study in North-East Netherlands. Int J Colorectal Dis. 2015;30:205–12.CrossRef Elferink MAG, de Jong KP, Klaase JM, et al. Metachronous metastases from colorectal cancer: a population-based study in North-East Netherlands. Int J Colorectal Dis. 2015;30:205–12.CrossRef
4.
Zurück zum Zitat Jayne DG, Fook S, Loi C, Seow-Choen F. Peritoneal carcinomatosis from colorectal cancer. Br J Surg. 2002;89(12):1545–50.CrossRef Jayne DG, Fook S, Loi C, Seow-Choen F. Peritoneal carcinomatosis from colorectal cancer. Br J Surg. 2002;89(12):1545–50.CrossRef
5.
Zurück zum Zitat Glehen O, Osinsky D, Beaujard AC, et al. Natural history of peritoneal carcinomatosis from nongynecologic malignancies. Surg Oncol Clin N Am. 2003;12(3):729–39.CrossRef Glehen O, Osinsky D, Beaujard AC, et al. Natural history of peritoneal carcinomatosis from nongynecologic malignancies. Surg Oncol Clin N Am. 2003;12(3):729–39.CrossRef
6.
Zurück zum Zitat Sugarbaker PH, Cunliffe WJ, Belliveau J, et al. Rationale for integrating early postoperative intraperitoneal chemotherapy into the surgical treatment of gastrointestinal cancer. Semin Oncol. 1989;16(4 Suppl 6):83–97.PubMed Sugarbaker PH, Cunliffe WJ, Belliveau J, et al. Rationale for integrating early postoperative intraperitoneal chemotherapy into the surgical treatment of gastrointestinal cancer. Semin Oncol. 1989;16(4 Suppl 6):83–97.PubMed
7.
Zurück zum Zitat Minsky BD, Mies C, Rich TA, et al. Potentially curative surgery of colon cancer: patterns of failure and survival. J Clin Oncol. 1988;6(1):106–18.CrossRef Minsky BD, Mies C, Rich TA, et al. Potentially curative surgery of colon cancer: patterns of failure and survival. J Clin Oncol. 1988;6(1):106–18.CrossRef
8.
Zurück zum Zitat Glehen O, Kwiatkowski F, Sugarbaker PH, et al. Cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for the management of peritoneal carcinomatosis from colorectal cancer: a multi-institutional study. J Clin Oncol. 2004;22(16):3284–92.CrossRef Glehen O, Kwiatkowski F, Sugarbaker PH, et al. Cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for the management of peritoneal carcinomatosis from colorectal cancer: a multi-institutional study. J Clin Oncol. 2004;22(16):3284–92.CrossRef
9.
Zurück zum Zitat Esquivel J, Elias D, Baratti D, et al. Consensus statement on the locoregional treatment of colorectal cancer with peritoneal dissemination. J Surg Oncol. 2008;98(4):263–7.CrossRef Esquivel J, Elias D, Baratti D, et al. Consensus statement on the locoregional treatment of colorectal cancer with peritoneal dissemination. J Surg Oncol. 2008;98(4):263–7.CrossRef
10.
Zurück zum Zitat Cortes-Guiral D, Elias D, Cascales-Campos PA, et al. Second-look surgery plus hyperthermic intraperitoneal chemotherapy for patients with colorectal cancer at high risk of peritoneal carcinomatosis: does it really save lives? World J Gastroenterol. 2017;23(3):377–81.CrossRef Cortes-Guiral D, Elias D, Cascales-Campos PA, et al. Second-look surgery plus hyperthermic intraperitoneal chemotherapy for patients with colorectal cancer at high risk of peritoneal carcinomatosis: does it really save lives? World J Gastroenterol. 2017;23(3):377–81.CrossRef
11.
Zurück zum Zitat Tomlinson JS, Jarnagin WR, DeMatteo RP, et al. Actual 10-year survival after resection of colorectal liver metastases defines cure. J Clin Oncol. 2007;25(29):4575–80.CrossRef Tomlinson JS, Jarnagin WR, DeMatteo RP, et al. Actual 10-year survival after resection of colorectal liver metastases defines cure. J Clin Oncol. 2007;25(29):4575–80.CrossRef
12.
Zurück zum Zitat Elias D, Lefevre JH, Chevalier J, et al. Complete cytoreductive surgery plus intraperitoneal chemohyperthermia with oxaliplatin for peritoneal carcinomatosis of colorectal origin. J Clin Oncol. 2009;27:681–5.CrossRef Elias D, Lefevre JH, Chevalier J, et al. Complete cytoreductive surgery plus intraperitoneal chemohyperthermia with oxaliplatin for peritoneal carcinomatosis of colorectal origin. J Clin Oncol. 2009;27:681–5.CrossRef
13.
Zurück zum Zitat Glehen O, Gilly FN, Boutitie F, et al. Toward curative treatment of peritoneal carcinomatosis from nonovarian origin by cytoreductive surgery combined with perioperative intraperitoneal chemotherapy: a multi-institutional study of 1290 patients. Cancer. 2010;116:5608–18.CrossRef Glehen O, Gilly FN, Boutitie F, et al. Toward curative treatment of peritoneal carcinomatosis from nonovarian origin by cytoreductive surgery combined with perioperative intraperitoneal chemotherapy: a multi-institutional study of 1290 patients. Cancer. 2010;116:5608–18.CrossRef
14.
Zurück zum Zitat Passot G, Vaudoyer D, Cotte E, et al. Progression following neoadjuvant systemic chemotherapy may not be a contraindication to a curative approach for colorectal carcinomatosis. Ann Surg. 2012;256:125–9.CrossRef Passot G, Vaudoyer D, Cotte E, et al. Progression following neoadjuvant systemic chemotherapy may not be a contraindication to a curative approach for colorectal carcinomatosis. Ann Surg. 2012;256:125–9.CrossRef
15.
Zurück zum Zitat Gelli M, Huguenin J, Cerebelli C, et al. Strategies to prevent peritoneal carcinomatosis arising from colorectal cancer. Future Med. 2017;13:907–18. Gelli M, Huguenin J, Cerebelli C, et al. Strategies to prevent peritoneal carcinomatosis arising from colorectal cancer. Future Med. 2017;13:907–18.
16.
Zurück zum Zitat Serrano Del Moral Á, Pérez Viejo E, Manzanedo Romero I, et al. Cirugía de second look más HIPEC en pacientes sin evidencia de recidiva con alto riesgo de desarrollar carcinomatosis tras resección de cáncer colorrectal. Cir Esp. 2018;96(2):97–102.CrossRef Serrano Del Moral Á, Pérez Viejo E, Manzanedo Romero I, et al. Cirugía de second look más HIPEC en pacientes sin evidencia de recidiva con alto riesgo de desarrollar carcinomatosis tras resección de cáncer colorrectal. Cir Esp. 2018;96(2):97–102.CrossRef
17.
Zurück zum Zitat Baratti D, Kusamura S, Iusco D, et al. Hyperthermic intraperitoneal Chemotherapy (HIPEC) at the time of primary curative surgery in patients with colorectal cancer at high risk for metachronous peritoneal metastases. Ann Surg Oncol. 2017;24(1):167–75.CrossRef Baratti D, Kusamura S, Iusco D, et al. Hyperthermic intraperitoneal Chemotherapy (HIPEC) at the time of primary curative surgery in patients with colorectal cancer at high risk for metachronous peritoneal metastases. Ann Surg Oncol. 2017;24(1):167–75.CrossRef
18.
Zurück zum Zitat Elias D, Honoré C, Dumont F, et al. Results of systematic second look surgery plus HIPEC in asymptomatic patients presenting a high risk of developing colorectal peritoneal carcinomatosis. Ann Surg. 2011;254:289–93.CrossRef Elias D, Honoré C, Dumont F, et al. Results of systematic second look surgery plus HIPEC in asymptomatic patients presenting a high risk of developing colorectal peritoneal carcinomatosis. Ann Surg. 2011;254:289–93.CrossRef
19.
Zurück zum Zitat Honoré C, Goéré D, Souadka A, et al. Definition of patients presenting a high risk of developing peritoneal carcinomatosis after curative surgery for colorectal cancer: a systematic review. Ann Surg Oncol. 2013;20:183–92.CrossRef Honoré C, Goéré D, Souadka A, et al. Definition of patients presenting a high risk of developing peritoneal carcinomatosis after curative surgery for colorectal cancer: a systematic review. Ann Surg Oncol. 2013;20:183–92.CrossRef
20.
Zurück zum Zitat Klaver CE, et al. Adjuvant hyperthermic intraperitoneal chemotherapy (HIPEC) in patients with colon cancer at high risk of peritoneal carcinomatosis; the COLOPEC randomized multicentre trial. BMC Cancer. 2015;15:428–37.CrossRef Klaver CE, et al. Adjuvant hyperthermic intraperitoneal chemotherapy (HIPEC) in patients with colon cancer at high risk of peritoneal carcinomatosis; the COLOPEC randomized multicentre trial. BMC Cancer. 2015;15:428–37.CrossRef
21.
Zurück zum Zitat Hompes D, Tiek J, Wolthuis A. HIPEC in T4a colon cancer: a defendable treatment to improve the oncologic outcome? Ann Oncol. 2012;23:3123–9.CrossRef Hompes D, Tiek J, Wolthuis A. HIPEC in T4a colon cancer: a defendable treatment to improve the oncologic outcome? Ann Oncol. 2012;23:3123–9.CrossRef
22.
Zurück zum Zitat Folprecht G, Köhne C, Lutz M, et al. Systemic chemotherapy in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res. 2007;134:425–40.PubMed Folprecht G, Köhne C, Lutz M, et al. Systemic chemotherapy in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res. 2007;134:425–40.PubMed
23.
Zurück zum Zitat Van Santvoort HC, Braam HJ, Spekreijse KR, et al. Peritoneal carcinomatosis in t4 colorectal cancer: occurrence and risk factors. Ann Surg Oncol. 2014;21(5):1686–91.CrossRef Van Santvoort HC, Braam HJ, Spekreijse KR, et al. Peritoneal carcinomatosis in t4 colorectal cancer: occurrence and risk factors. Ann Surg Oncol. 2014;21(5):1686–91.CrossRef
24.
Zurück zum Zitat Segelman J, Granath F, Holm T, et al. Incidence, prevalence and risk factors for peritoneal carcinomatosis from colorectal cancer. Br J Surg. 2012;99(5):699–705.CrossRef Segelman J, Granath F, Holm T, et al. Incidence, prevalence and risk factors for peritoneal carcinomatosis from colorectal cancer. Br J Surg. 2012;99(5):699–705.CrossRef
25.
Zurück zum Zitat Ripley RT, Davis JL, Kemp CD, et al. Prospective randomized trial evaluating mandatory second look surgery with HIPEC and CRS vs. standard of care in patients at high risk of developing colorectal peritoneal metastases. Trials. 2010;11:62.CrossRef Ripley RT, Davis JL, Kemp CD, et al. Prospective randomized trial evaluating mandatory second look surgery with HIPEC and CRS vs. standard of care in patients at high risk of developing colorectal peritoneal metastases. Trials. 2010;11:62.CrossRef
26.
Zurück zum Zitat Goere D, Glehen O, Quenet F, et al. Results of a randomized phase 3 study evaluating the potential benefit of a second look surgery plus HIPEC in patients at high risk of developing colorrectal peritoneal disease. J Clin Oncol. 2018;36(15):3531.CrossRef Goere D, Glehen O, Quenet F, et al. Results of a randomized phase 3 study evaluating the potential benefit of a second look surgery plus HIPEC in patients at high risk of developing colorrectal peritoneal disease. J Clin Oncol. 2018;36(15):3531.CrossRef
27.
Zurück zum Zitat Quenet F, Elias D, Roca L, et al. A UNICANCER phase III trial of Hyperthermic intra-peritoneal chemotherapy (HIPEC) for colorrectal peritoneal carcinomatosis (PC): PRODIGE 7 (NCT 00769405). Quenet F, Elias D, Roca L, et al. A UNICANCER phase III trial of Hyperthermic intra-peritoneal chemotherapy (HIPEC) for colorrectal peritoneal carcinomatosis (PC): PRODIGE 7 (NCT 00769405).
28.
Zurück zum Zitat Arjona A, Barrios P, Boldo-Roda E, et al. HIPECT4: Multicentre randomized clinical trial to evaluate safety and efficacy of HIPEC with Mitomycin C used during surgery for the treatment of locally advanced colorectal carcinoma. BMC. 2018;18:183. Arjona A, Barrios P, Boldo-Roda E, et al. HIPECT4: Multicentre randomized clinical trial to evaluate safety and efficacy of HIPEC with Mitomycin C used during surgery for the treatment of locally advanced colorectal carcinoma. BMC. 2018;18:183.
29.
Zurück zum Zitat Proactive management of endoperitoneal spread in colonic cancer. PROMENADE trial (NCT 02974556). Proactive management of endoperitoneal spread in colonic cancer. PROMENADE trial (NCT 02974556).
30.
Zurück zum Zitat Trial evaluating surgery with hyperthermic intraperitoneal chemotherapy (HIPEC) in treating patients with a high risk of developing colorectal peritoneal carcinomatosis (NCT 02179489). Trial evaluating surgery with hyperthermic intraperitoneal chemotherapy (HIPEC) in treating patients with a high risk of developing colorectal peritoneal carcinomatosis (NCT 02179489).
Metadaten
Titel
Prophylactic HIPEC in pT4 Colon Tumors: Proactive Approach or Overtreatment?
verfasst von
Nerea Borda Arrizabalaga, MD
José María Enriquez Navascués, PhD
Garazi Elorza Echaniz, MD
Yolanda Saralegui Ansorena, PhD
Carlos Placer Galán, PhD
Xabier Arteaga Martín, PhD
Leyre Velaz Pardo, MD
Publikationsdatum
29.10.2019
Verlag
Springer International Publishing
Erschienen in
Annals of Surgical Oncology / Ausgabe 4/2020
Print ISSN: 1068-9265
Elektronische ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-019-07970-z

Weitere Artikel der Ausgabe 4/2020

Annals of Surgical Oncology 4/2020 Zur Ausgabe

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.