1,895
Views
17
CrossRef citations to date
0
Altmetric
Reviews

Treatment of peritoneal metastases from small bowel adenocarcinoma

, , &
Pages 571-578 | Received 04 Nov 2016, Accepted 27 Nov 2016, Published online: 30 Jun 2017

Abstract

Background/purpose: Peritoneal metastases (PM) affect approximately one third of patients with metastatic small bowel adenocarcinoma (SBA). Treatment options are (1) systemic therapy ± palliative surgery and (2) cytoreductive surgery with intraperitoneal chemotherapy (CRS + IPC). Due to scarce evidence, PM from SBA represents a therapeutic challenge. This narrative review summarised and discussed the evidence that investigated available treatment options.

Methods: Studies were discussed if they investigated first line systemic therapy for advanced SBA or CRS + IPC for PM from SBA. Extracted outcomes were objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), disease-free survival (DFS), overall survival (OS), and grade III–V toxicity/morbidity.

Results: Eighteen studies (15 observational, 3 phase II) that investigated systemic therapy and six observational studies that investigated CRS + IPC were reviewed. In studies that investigated systemic therapy, ORR, DCR, median PFS, median OS, and grade III–V toxicity ranged from 6% to 50%, 50% to 90%, 3 to 11 months, 8 to 20 months, and 10% to 68%, respectively. Fluoropyrimidine–oxaliplatin revealed favourable survival outcomes compared to fluoropyrimidine–irinotecan, fluoropyrimidine–cisplatin, fluoropyrimidine monotherapy, and other regimens. In studies that investigated CRS + IPC, median DFS, median OS, and grade III–V morbidity ranged from 10 to 12 months, 16 to 47 months, and 12% to 35%, respectively.

Conclusion: Based on available evidence, fluoropyrimidine–oxaliplatin should be regarded as optimal first line systemic treatment. In selected patients, CRS + IPC appears safe and may be more effective than systemic therapy as single treatment. Future studies should evaluate survival and morbidity of CRS + IPC in larger cohorts, as well as the value of chemotherapy with targeted agents in metastatic SBA with subgroup analysis for PM from SBA.

Introduction

Approximately 5% of all gastrointestinal malignancies arises from the small bowel [Citation1]. Small bowel adenocarcinoma (SBA) accounts for 30%–45% of all small bowel malignancies [Citation2–6], with estimated age-standardised incidences of 0.5 per 100 000 in 1999 and 0.7 per 100 000 in 2013 [Citation7]. SBA is mostly diagnosed in an advanced stage due to a low index of suspicion, vagueness of symptoms, difficult endoscopic access, and poor detection by radiologic imaging. As a result, approximately one third of patients with SBA present with stage IV disease at the time of diagnosis [Citation2,Citation3,Citation7–9]. Peritoneal metastases (PM) affect 25%–50% with stage IV SBA and are more common in SBA arising from the jejunum or ileum than SBA arising from the duodenum [Citation7,Citation8,Citation10–18] ().

Table 1. Percentage of patients with PM from SBA.

Systemic therapy has been increasingly used for stage IV SBA [Citation7]. This is based on scarce evidence obtained from several observational studies and small phase II trials in patients with advanced SBA [Citation8–27]. The extrapolation of this evidence to PM from SBA may be questioned, since clinical studies in colorectal cancer (CRC) demonstrated that PM are relatively resistant to systemic therapy compared to other isolated metastatic sites [Citation28–32].

A locoregional treatment option for PM from SBA is cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (CRS + HIPEC). This treatment modality is gaining acceptance as standard of care for pseudomyxoma peritonei, peritoneal mesothelioma, and selected patients with PM from CRC [Citation33,Citation34]. CRS + HIPEC may also be beneficial in PM from SBA, since molecular characteristics and carcinogenesis of SBA and colorectal adenocarcinoma are roughly similar [Citation35,Citation36]. Evidence to support this hypothetical benefit is based on several observational studies [Citation37–43].

Due to the rarity of PM from SBA and the paucity of data regarding systemic and surgical treatment, PM from SBA represent a major therapeutic challenge for surgeons and medical oncologists. Therefore, this narrative review aims to summarise and discuss studies that investigated available treatment options in order to provide treatment recommendations and future research directions.

Methods

Study eligibility

All studies that included patients who received first line systemic therapy for advanced SBA or CRS + IPC for PM from SBA were discussed in this review. Studies were considered ineligible if they were published >20 years ago or included ≤3 patients.

Data collection and outcome parameters

If reported, the following data were collected for each study by using a standardised form: year of publication, years of patient inclusion, study design, number of patients, primary tumour location, locally advanced or metastatic disease, percentage of patients who received first line systemic therapy, systemic therapy regimens, percentage of patients who received (neo)adjuvant systemic therapy, procedure related characteristics of CRS + IPC (e.g. IPC technique, IPC drugs, completeness of cytoreduction score, peritoneal cancer index).

If reported, the following outcome parameters were extracted by using a standardised form: objective response rate (ORR) in %, disease control rate (DCR) in %, median progression-free survival (PFS) in months, median disease-free survival (DFS) in months, median overall survival (OS) in months, and grade III–V morbidity/toxicity in %. Results from chi-square test (X2), log-rank test, and multivariable regression analyses were extracted if studies compared predefined outcome parameters between different interventions within the study.

ORR and DCR were based on either the Response Criteria in Solid Tumours (RECIST) or the World Health Organisation (WHO) response measurement criteria [Citation44–46]. Toxicity and morbidity were graded according to either the National Cancer Institute Common Toxicity Criteria, National Cancer Institute Common Terminology Criteria for Adverse Events, or Clavien-Dindo [Citation47–49].

Results

Twenty-four studies were discussed. Supplementary Appendix 1 demonstrates the data collection form for all studies.

Systemic therapy

Three phase II studies () and 15 observational studies reported predefined outcome parameters in patients who received first line systemic for advanced SBA () [Citation8–14,Citation16–19,Citation21–27]. Five observational studies were non-comparative and reported predefined outcomes parameters of their total cohorts () [Citation11,Citation19,Citation23,Citation24,Citation26]. Ten observational studies were comparative. Of these studies, four compared predefined outcome parameters between different systemic therapy regimens () [Citation10,Citation12,Citation14,Citation17], and six compared survival outcomes in patients who received systemic therapy vs. no systemic therapy () [Citation8,Citation9,Citation16,Citation22,Citation25,Citation27].

Table 2. Studies that investigate ORR, DCR, PFS, OS, or grade III–V toxicity (NCICTC) in patients who receive first line systemic therapy for advanced SBA.

Tumour response

Twelve studies reported ORR or DCR. ORR ranged from 6% to 50% and DCR ranged from 50% to 90% between studies [Citation10,Citation11,Citation13,Citation14,Citation18,Citation19,Citation22–27].

In phase II studies, ORR was 49% after 5-fluorouracil (5-FU) with leucovorin and oxaliplatin (FOLFOX) and 50% after capecitabine and oxaliplatin (CAPOX) [Citation13,Citation18]. DCR was 85% after FOLFOX and 90% after CAPOX [Citation13,Citation18].

In non-comparative observational studies, ORR and DCR were 6%–11% and 56% after 5-FU monotherapy [Citation22,Citation24], 13% and 75% after irinotecan and cisplatin [Citation23], 32% and 62% after a fluoropyrimidine and oxaliplatin [Citation26], and 50% and 67% after modern doublet and triplet systemic therapy regimens, respectively [Citation27]. Other studies that used various regimens reported an ORR and DCR ranging from 28% to 37% and 50% to 79%, respectively [Citation10,Citation11,Citation19,Citation25].

In a French single-centre comparative observational study (n = 20), ORR after 5-FU and carboplatin, 5-FU and cisplatin, and FOLFOX was 0%, 20%, and 33%, respectively (no statistical analysis) [Citation10]. In a larger American single-centre comparative observational study (n = 80), a higher ORR was found after 5-FU with a platinum compound vs. 5-FU monotherapy/other regimens (46% vs. 16%; X2: p = 0.01) [Citation12]. In a French multicentre comparative observational study (n = 93), ORR and DCR after 5-FU monotherapy, 5-FU with leucovorin and irinotecan (FOLFIRI), 5-FU and cisplatin, and FOLFOX were 0% and 50%, 9% and 73%, 31% and 69%, and 34% and 79%, respectively (X2: p = 0.18) [Citation14]. In a Japanese multicentre comparative observational study (n = 132), ORR after 5-FU monotherapy, FOLFIRI, 5-FU and cisplatin, FOLFOX, and other regimens was 20%, 25%, 38%, 42% and 21%, respectively (no statistical analysis) [Citation17].

Progression-free survival

Twelve studies reported median PFS, which ranged from 3 to 11 months between studies [Citation10,Citation12–14,Citation16,Citation18,Citation19,Citation21,Citation23–26].

In phase II studies, median PFS was 8 months after FOLFOX [Citation18], 11 months after CAPOX [Citation13], and 5 months after 5-FU, mitomycin C, and doxorubicin [Citation21].

In non-comparative observational studies, median PFS was 3 months after 5-FU monotherapy [Citation24], 5 months after irinotecan and cisplatin [Citation23], 6–8 months after fluoropyrimidines and a platinum compound [Citation10,Citation26], and 4–8 months in studies that used various regimens [Citation19,Citation25].

In a Korean single-centre comparative observational study, median PFS was 6 months in patients who received chemotherapy compared to 1 month in patients who did not receive chemotherapy (log-rank: p < 0.01) [Citation16]. In the American single-centre study, median PFS after 5-FU with and without a platinum compound was 9 months vs. 4 months, respectively (log-rank: p < 0.01) [Citation12]. In the French multicentre study, median PFS after 5-FU monotherapy, FOLFIRI, 5-FU and cisplatin, and FOLFOX was 8 months, 6 months, 5 months, and 7 months, respectively (log-rank: p = 0.16) [Citation14]. In this study, median PFS was shorter after 5-FU and cisplatin than after FOLFOX on univariable (log-rank: p = 0.02) and multivariable analysis (HR: 2.25 [95%CI 1.12–4.50]) [Citation14]. In the Japanese multicentre study, median PFS after 5-FU monotherapy, FOLFIRI, 5-FU and cisplatin, FOLFOX, and other regimens were 5 months, 6 months, 4 months, 8 months, and 3 months, respectively [Citation17]. In this study, median PFS was higher after FOLFOX vs. 5-FU monotherapy on univariable (log-rank p = 0.03) and multivariable analysis (HR 0.48 [95%CI 0.23–0.84]), whereas no statistically significant difference in PFS was found between 5-FU monotherapy and FOLFIRI, 5-FU and cisplatin, or other regimens [Citation17].

Overall survival

Seventeen studies reported median OS, which ranged from 8 to 20 months between studies [Citation8–14,Citation16,Citation18,Citation19,Citation21,Citation22–27].

In phase II studies, median OS was 15 months after FOLFOX [Citation18], 20 months after CAPOX [Citation13], and 8 months after 5-FU, mitomycin C, and doxorubicin [Citation21].

In non-comparative observational studies, median OS was 12 months after 5-FU monotherapy [Citation24], 17 months after irinotecan and cisplatin [Citation23], 14 months after fluoropyrimidines and a platinum compound [Citation10,Citation26], and 11–13 months in studies that used various regimens [Citation11,Citation19].

Six comparative studies revealed higher median OS in patients who received chemotherapy (12–16 months) vs. patients who did not receive chemotherapy (2–8 months) (log-rank: p < 0.05 in all studies) [Citation8,Citation9,Citation16,Citation22,Citation25,Citation27]. In the American single-centre study, median OS after 5-FU with and without a platinum compound was 15 vs. 12 months, respectively (log-rank: p = 0.10) [Citation12]. In the French multicentre study, median OS after 5-FU monotherapy, FOLFIRI, 5-FU and cisplatin, and FOLFOX was 14 months, 11 months, 9 months and 18 months, respectively (log-rank p = 0.25) [Citation14]. In this study, median OS was shorter after 5-FU and cisplatin compared to FOLFOX on univariable (log-rank: p = 0.04) and multivariable analysis (HR: 2.75 [95%CI 1.18–6.41]) [Citation14]. In the Japanese multicentre study, median OS after 5-FU monotherapy, FOLFIRI, 5-FU and cisplatin, FOLFOX, and other regimens was 14 months, 9 months, 13 months, 22 months, and 8 months, respectively [Citation17]. In this study, median OS did not differ between FOLFOX and 5-FU monotherapy on univariable analysis (log-rank: p = 0.16). However, median OS was higher after FOLFOX vs. 5-FU monotherapy on multivariable analysis (HR: 0.45 [95%CI 0.23–0.88]), whereas no difference in OS was found between 5-FU monotherapy and FOLFIRI, 5-FU and cisplatin, or other regimens [Citation17].

Grade III–V toxicity

Six studies reported grade III–V toxicity, which ranged from 10% to 68% between studies [Citation10,Citation14,Citation18,Citation19,Citation21,Citation24].

In phase II studies, grade III–V toxicity was 39% with FOLFOX [Citation18], and 68% with 5-FU, mitomycin C, and doxorubicin [Citation21].

In non-comparative observational studies, grade III–V toxicity was 10% with 5-FU monotherapy [Citation24], and 25% in a study that used various regimens [Citation19].

In the French single-centre study, grade III–V toxicity of 5-FU and carboplatin, 5-FU and cisplatin, and FOLFOX were 0%, 53%, and 33%, respectively (no statistical analysis) [Citation10]. In the French multicentre study, grade III–V toxicity of 5-FU monotherapy, FOLFIRI, 5-FU and cisplatin, and FOLFOX was 0%, 39%, 75%, and 46%, respectively [Citation14]. In this study, grade III–V toxicity was higher with 5-FU and cisplatin compared to other regimens (X2: p < 0.01) [Citation14].

Cytoreductive surgery with intraperitoneal chemotherapy

Six observational studies reported predefined outcome parameters in patients who underwent CRS + IPC for PM from SBA () [Citation37,Citation39–43]. The percentage of patients who received neoadjuvant and adjuvant systemic therapy ranged from 14% to 90% and 29% to 57% between studies, respectively [Citation39–43].

Disease-free and overall survival

Two studies reported median DFS, which was 12 months after CRS + HIPEC with mitomycin C [Citation42] and 10 months in a study that used various IPC techniques and drugs [Citation39].

Table 3. Observational studies that investigate DFS, OS, and grade III–V morbidity in patients who undergo cytoreductive surgery with intraperitoneal chemotherapy for peritoneal metastases arising from SBA.

Six studies reported median OS measured from CRS + IPC, which ranged from 16 to 47 months between studies [Citation37,Citation39–43]. Median OS was 16 months after CRS + HIPEC with mitomycin C and early postoperative intraperitoneal chemotherapy (EPIC) with 5-FU [Citation37], 18–36 months after CRS + HIPEC with mitomycin C [Citation41–43], and 25–47 months in studies that used various IPC techniques and drugs [Citation39,Citation40]. Median OS measured from diagnosis was reported in two studies that used CRS + HIPEC with mitomycin C and ranged from 37 to 51 months [Citation41,Citation43].

Grade III–V morbidity

Five studies reported grade III–V morbidity, which ranged from 12% to 35% between studies [Citation39–43]. Grade III–V morbidity was 12%–26% after CRS + HIPEC with mitomycin C [Citation41–43], and 29%–35% in studies that used various IPC techniques and drugs [Citation39,Citation40].

Discussion

Based on results of phase II and comparative observational studies, first line systemic treatment with a fluoropyrimidine and oxaliplatin seems to achieve favourable survival outcomes and at least equal toxicity and tumour response outcomes compared to regimens with a fluoropyrimidine and irinotecan or cisplatin. In selected patients with PM from SBA, CRS + IPC with or without (neo)adjuvant systemic therapy seems safe and may be more effective than systemic therapy as single treatment, with promising survival and morbidity outcomes.

Systemic therapy

In daily practice, systemic therapy is the most common treatment option for PM from SBA. In the absence of prospective randomised studies, there is no consensus on the preferred first line regimen in metastatic SBA, even though observational studies in this review point towards superior results of a fluoropyrimidine with oxaliplatin. Therefore, information obtained from observational and phase II studies in metastatic SBA may be complemented with evidence derived from metastatic CRC, which has similar molecular characteristics and carcinogenesis pathways [Citation35,Citation36].

In metastatic CRC, a fluoropyrimidine with oxaliplatin has been standard of care for many years [Citation50]. In 2011, a phase II study revealed a promising median OS of 21.5 months in patients who received FOLFOX for metastatic CRC with PM [Citation51]. More recently, addition of targeted agents to combination chemotherapy significantly improved survival of metastatic CRC [Citation52]. The role of targeted agents in metastatic SBA is less clear. However, the high expression of vascular endothelial growth factor (VEGF) A (96%) and epidermal growth factor receptor (EGFR) (71%) in SBA may suggest a role for treatment with targeted agents such as bevacizumab or cetuximab [Citation53].

Interestingly, addition of bevacizumab to chemotherapy improved survival of PM from CRC [Citation54]. Specifically for peritoneal tumour depositions, VEGF is known to have a role in tumour related angiogenesis and formation of ascites [Citation55]. Therefore, VEGF inhibitors may be of value as addition to chemotherapy for the treatment of PM from various gastrointestinal origins, including SBA. However, the role of VEGF inhibitors in this setting needs to be further clarified.

Limitations of available evidence

None of the studies that investigated systemic therapy for advanced SBA provided a subgroup analysis of patients with PM. In a recently published pooled subgroup analysis of 14 prospective randomised controlled trials in metastatic CRC, patients with PM demonstrated shorter OS compared to patients with other isolated sites of metastases [Citation32]. However, presence of PM was no predictor of survival in studies in this review [Citation12,Citation14–16]. Nevertheless, the absence of this subgroup analysis impedes the extrapolation of results of systemic therapy for patients with any form of advanced SBA to patients with PM from SBA.

Furthermore, all studies in this review included patients with SBA arising from duodenum, ileum, and jejunum. However, duodenal adenocarcinomas arise mostly retroperitoneal and are known for a more advanced disease at presentation, worse prognosis, and a different metastatic pattern with less peritoneal seeding [Citation7]. Therefore, it could be questioned whether these malignancies should be regarded as one disease entity in oncological studies.

Finally, given the large inclusion periods of studies in this review, several non-comparative studies only used conventional regimens [Citation21,Citation22,Citation24], and other studies that used both conventional and modern regimens reported outcomes in their total cohort [Citation10,Citation11,Citation16,Citation19,Citation25]. This interventional heterogeneity impedes extrapolation of results to patients who are treated in the era of modern systemic therapy regimens.

Future research directions

The value of targeted agents in metastatic SBA is currently under investigation in a phase IB (ClinicalTrials.gov: NCT00987766) and two phase II studies (ClinicalTrials.gov: NCT01202409, NCT01208103). Ideally, future studies that focus on systemic therapy for metastatic SBA should provide subgroup analyses for PM from SBA, in order to facilitate a more reliable comparison of survival outcomes between systemic therapy as single treatment and CRS + IPC. Based on the current review, patients with PM from SBA may be treated with a fluoropyrimidine with oxaliplatin until results of on-going trials become available. Since there is only one observational study that investigated second line chemotherapy (FOLFIRI) in advanced SBA [Citation15], future studies are needed to identify the optimal second line regimen.

Cytoreductive surgery with intraperitoneal chemotherapy

In selected patients with PM from SBA, CRS + IPC seems to be an attractive treatment option. Selection criteria include absence of unresectable systemic disease, achievability of a complete CRS, and a physical status that allows a major surgical procedure. In the two most recent studies by van Oudheusden et al. [Citation42] and Liu et al. [Citation43], survival and morbidity outcomes were comparable with those after CRS + HIPEC for PM from CRC [Citation34]. The median OS of 31–36 months in these studies is compatible with the pursuit of at least 30 months after CRS + HIPEC in PM from CRC as defined by the Peritoneal Surface Oncology Group International [Citation56]. Given these promising survival results, efforts should be made to increase awareness regarding this treatment option. The low number of patients with PM from SBA who were treated with CRS + HIPEC demonstrates that this awareness is currently lacking. In the Netherlands, 16 patients were treated with CRS + HIPEC for PM from SBA over a 10 year time period [Citation42], while the number of diagnosed patients was significantly higher. A Dutch epidemiological study revealed that 167 patients were diagnosed with synchronous PM from SBA between 1999 and 2013 [Citation7]. Approximately 60% of these patients presented with isolated PM without concurrent systemic metastases (Dutch Cancer Registry), and are therefore potential candidates for CRS + HIPEC. These results underestimate the true number of patients with isolated PM from SBA, since data on metachronous PM from SBA are not available. The difference between the number of patients diagnosed with isolated PM from SBA and the number of patients who underwent CRS + HIPEC may indicate underuse of CRS + HIPEC for potentially eligible patients.

Limitations of available evidence

All studies that investigated CRS + HIPEC for PM from SBA have an observational design and no prospective studies on this topic have been conducted. Due to large inclusion periods within studies and different inclusion periods across studies, heterogeneity in interventions both within and across studies was observed. Several studies only used CRS + HIPEC [Citation41–43], whereas other studies mainly used CRS + EPIC [Citation37,Citation39,Citation40]. Moreover, (neo)adjuvant treatment varied across studies from mainly adjuvant [Citation39,Citation40,Citation42], to mainly neoadjuvant [Citation43], or both [Citation41]. Since both HIPEC or EPIC and neoadjuvant or adjuvant systemic therapy appear to have a different influence on morbidity and survival in CRC [Citation57–64], this heterogeneity may have biased reported outcomes.

Besides interventional heterogeneity, heterogeneity was also observed in included patients. Some studies only included patients with a complete cytoreduction [Citation38,Citation39] and a low peritoneal cancer index (PCI) [Citation39,Citation41], whereas other studies also included patients with an incomplete cytoreduction [Citation40–42] and a high PCI [Citation38,Citation42]. Since completeness of cytoreduction and PCI are known to be the most important prognostic factors for survival in CRC [Citation65], this heterogeneity may have influenced survival outcomes.

Finally, the small number of included patients impedes statistical analysis for prognostic variables that predict and influence survival and morbidity.

Future research directions

Limitations of available evidence stress the need for higher quality studies in order to confirm the promising results of CRS + HIPEC in selected patients with PM from SBA. Given the rarity of the disease, it is unlikely that this evidence will ever be provided by a prospective randomised study. In order to increase the number of patients in observational studies, a multi-institutional data registry on PM from SBA was established during the 9th International Congress on Peritoneal Surface Malignancies in 2014. Forthcoming results of this data registry will provide more insight in (1) survival and morbidity outcomes in a larger cohort and (2) prognostic patient, tumour and treatment related variables that predict and influence survival and morbidity.

If a patient is no candidate for CRS + HIPEC due to extensive peritoneal disease or a poor performance status, pressurised intraperitoneal aerosol chemotherapy (PIPAC) may be a future treatment option for symptomatic peritoneal disease [Citation66]. Preliminary results of this innovative locoregional treatment modality, which may be combined with systemic therapy in case of concurrent systemic disease, revealed interesting peritoneal tumour regression in end-stage PM arising from gastric [Citation67], colorectal [Citation68], and ovarian cancer [Citation69], without quality of life deterioration [Citation70]. Several on-going studies are currently investigating this technique for PM from various primary malignancies (ClinicalTrials.gov: NCT02475772, NCT01809379, NCT01854255, NCT02320448, NCT02735928, NCT02604784). However, by our knowledge, no PIPAC has been performed in patients with PM from SBA thus far.

Conclusions

Based on available evidence, a fluoropyrimidine with oxaliplatin seems to be the optimal first line systemic therapy regimen for patients with PM from SBA. In selected patients, CRS + IPC appears safe and may be more effective than systemic therapy as single treatment. Future studies should evaluate survival and morbidity of CRS + IPC in larger cohorts, as well as the value of systemic chemotherapy with targeted agents in metastatic SBA with subgroup analysis for PM from SBA.

Supplemental material

Supplemental File

Download Zip (156.8 KB)

Disclosure statement

The authors report no declarations of interest.

References

  • Siegel RL, Miller KD, Jemal A. (2016). Cancer statistics, 2016. CA Cancer J Clin 66:7–30.
  • Howe JR, Karnell LH, Menck HR, et al. (1999). Adenocarcinoma of the small bowel: review of the National Cancer Data Base, 1985–1995. Cancer 86:2693–706.
  • Talamonti MS, Goetz LH, Rao S, Joehl RJ. (2002). Primary cancers of the small bowel: analysis of prognostic factors and results of surgical management. Arch Surg 137:564–70.
  • Lepage C, Bouvier AM, Manfredi S, et al. (2006). Incidence and management of primary malignant small bowel cancers: a well-defined French population study. Am J Gastroenterol 101:2826–32.
  • Hatzaras I, Palesty JA, Abir F, et al. (2007). Small-bowel tumors: epidemiologic and clinical characteristics of 1260 cases from the Connecticut tumor registry. Arch Surg 142:229–35.
  • Bilimoria KY, Bentrem DJ, Wayne JD, et al. (2009). Small bowel cancer in the United States: changes in epidemiology, treatment, and survival over the last 20 years. Ann Surg 249:63–71.
  • Legué LM, Bernards N, Gerritse SL, et al. (2016). Trends in incidence, treatment and survival of small bowel adenocarcinomas between 1999 and 2013: a population-based study in the Netherlands. Acta Oncol 55:1183–9.
  • Debaja BS, Suki D, Pro B, et al. (2004). Adenocarcinoma of the small bowel: presentation, prognostic factors, and outcome of 217 patients. Cancer 101:518–26.
  • Halfdanarson TR, McWilliams RR, Donohue JH, Quevedo JF. (2010). A single-institution experience with 491 cases of small bowel adenocarcinoma. Am J Surg 199:797–803.
  • Locher C, Malka D, Boige V, et al. (2005). Combination chemotherapy in advanced small bowel adenocarcinoma. Oncology 69:290–4.
  • Fishman PN, Pond GR, Moore MJ, et al. (2006). Natural history and chemotherapy effectiveness for advanced adenocarcinoma of the small bowel: a retrospective review of 113 cases. Am J Clin Oncol 29:225–31.
  • Overman MJ, Kopetz S, Wen S, et al. (2008). Chemotherapy with 5-fluorouracil and a platinum compound improves outcomes in metastatic small bowel adenocarcinoma. Cancer 113:2038–45.
  • Overman MJ, Varadhachary GR, Kopetz S, et al. (2009). Phase II study of capecitabine and oxaliplatin for advanced adenocarcinoma of the small bowel and ampulla of Vater. J Clin Oncol 27:2598–603.
  • Zaanan A, Costes L, Gauthier M, et al. (2010). Chemotherapy of advanced small-bowel adenocarcinoma: a multicenter AGEO study. Ann Oncol 21:1786–93.
  • Zaanan A, Gauthier M, Malka D, et al. (2011). Second-line chemotherapy with fluorouracil, leucovorin, and irinotecan (FOLFIRI regimen) in patients with advanced small bowel adenocarcinoma after failure of first-line platinum-based chemotherapy: a multicenter AGEO study. Cancer 117:1422–8.
  • Koo DH, Yun SC, Hong YS, et al. (2011). Systemic chemotherapy for treatment of advanced small bowel adenocarcinoma with prognostic factor analysis: retrospective study. BMC Cancer 11:205.
  • Tsushima T, Taguri M, Honma Y, et al. (2012). Multicenter retrospective study of 132 patients with unresectable small bowel adenocarcinoma treated with chemotherapy. Oncologist 17:1163–70.
  • Xiang XJ, Liu YW, Zhang L, et al. (2012). A phase II study of modified FOLFOX as first-line chemotherapy in advanced small bowel adenocarcinoma. Anticancer Drugs 23:561–6.
  • Crawley C, Ross P, Norman A, et al. (1998). The Royal Marsden experience of a small bowel adenocarcinoma treated with protracted venous infusion 5-fluorouracil. Br J Cancer 78:508–10.
  • Polyzos A, Kouraklis G, Giannopoulos A, et al. (2003). Irinotecan as salvage chemotherapy for advanced small bowel adenocarcinoma: a series of three patients. J Chemother 15:503–6.
  • Gibson MK, Holcroft CA, Kvols LK, Haller D. (2005). Phase II study of 5-fluorouracil, doxorubicin, and mitomycin C for metastatic small bowel adenocarcinoma. Oncologist 10:132–7.
  • Czaykowski P, Hui D. (2007). Chemotherapy in small bowel adenocarcinoma: 10-year experience of the British Columba Cancer Agency. Clin Oncol (R Coll Radiol) 19:143–9.
  • Ono M, Shirao K, Takashima A, et al. (2009). Combination chemotherapy with cisplatin and irinotecan in patients with adenocarcinoma of the small intestine. Gastric Cancer 11:201–5.
  • Suenaga M, Mizunuma N, Chin K, et al. (2009). Chemotherapy for small-bowel adenocarcinoma at a single institution. Surg Today 39:27–31.
  • Moon YW, Rha SY, Shin SJ, (2010). Adenocarcinoma of the small bowel at a single Korean institute: management and prognosticators. J Cancer Res Clin Oncol 136:387–94.
  • Zhang L, Wang LY, Deng YM, et al. (2011). Efficacy of the FOLFOX/CAPOX regimen for advanced small bowel adenocarcinoma: a three-center study from China. J Buon 16:689–96.
  • Khan K, Peckitt C, Sclafani F, et al. (2015). Prognostic factors and treatment outcomes in patients with Small Bowel Adenocarcinoma (SBA): the Royal Marsden Hospital (RMH) experience. BMC Cancer 15:15.
  • Assersohn L, Norman A, Cunningham D, et al. (1999). Influence of metastatic site as an additional predictor for response and outcome in advanced colorectal carcinoma. Br J Cancer 79:1800–5.
  • Kohne CH, Cunningham D, di Constanzo F, et al. (2002). Clinical determinants of survival in patients with 5-fluorouracil-based treatment for metastastic colorectal cancer: results of a multivariate analysis of 3825 patients. Ann Oncol 13:308–17.
  • Franko J, Shi Q, Goldman CD, et al. (2012). Treatment of colorectal peritoneal carcinomatosis with systemic chemotherapy: a pooled analysis of north central cancer treatment group phase III trials N9741 and N9841. J Clin Oncol 30:263–7.
  • Klaver YL, Simkens LH, Lemmens VE, et al. (2012). Outcomes of colorectal cancer patients with peritoneal carcinomatosis treated with chemotherapy with and without targeted therapy. Eur J Surg Oncol 38:617–23.
  • Franko J, Shi Q, Meyers JP, et al. (2016). Prognosis of patients with peritoneal metastatic colorectal cancer given systemic therapy: an analysis of individual patient data from prospective randomised trials from the Analysis and Research in Cancers of the Digestive System (ARCAD) database. Lancet Oncol 17:1709–19.
  • Elias D, Goéré D, Dumont F, et al. (2014). Role of hyperthermic intraoperative peritoneal chemotherapy in the management of peritoneal metastases. Eur J Cancer 50:332–40.
  • Baratti D, Kusamura S, Pietrantonio F, et al. (2016). Progress in treatments for colorectal cancer peritoneal metastases during the years 2010–2015. A systematic review. Crit Rev Oncol Hematol 100:209–22.
  • Aparicio T, Svrcek M, Zaanan A, et al. (2013). Small bowel adenocarcinoma phenotyping, a clinicobiological prognostic study. Br J Cancer 109:3057–66.
  • Zaaimi Y, Aparicio T, Laurent-Puig P, et al. (2016). Advanced small bowel adenocarcinoma: molecular characteristics and therapeutic perspectives. Clin Res Hepatol Gastroenterol 40:154–60.
  • Marchettini P, Sugarbaker PH. (2002). Mucinous adenocarcinoma of the small bowel with peritoneal seeding. Eur J Surg Oncol 28:19–23.
  • Jacks SP, Hundley JC, Shen P, et al. (2005) Cytoreductive surgery and intraperitoneal hyperthermic chemotherapy for peritoneal carcinomatosis from small bowel adenocarcinoma. J Surg Oncol 91:112–17.
  • Chua TC, Koh JL, Yan TD, et al. (2009). Cytoreductive surgery and perioperative intraperitoneal chemotherapy for peritoneal carcinomatosis from small bowel adenocarcinoma. J Surg Oncol 100:139–43.
  • Elias D, Glehen O, Pocard M, et al. (2010). A comparative study of complete cytoreductive surgery plus intraperitoneal chemotherapy to treat peritoneal dissemination from colon, rectum, small bowel, and nonpseudomyxoma appendix. Ann Surg 251:896–901.
  • Sun Y, Shen P, Stewart JH, et al. (2013). Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for peritoneal carcinomatosis from small bowel adenocarcinoma. Am Surg 79:644–8.
  • van Oudheusden TR, Lemmens VE, Braam HJ, et al. (2015). Peritoneal metastases from small bowel cancer: results of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in the Netherlands Surgery 157:1023–7.
  • Liu Y, Ishibashi H, Takeshita K, et al. (2016). Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for peritoneal dissemination from small bowel malignancy: results from a single specialized center. Ann Surg Oncol 23:1625–31.
  • Miller AB, Hoogstraten B, Staquet M, Winkler A. (1981). Reporting results of cancer treatment. Cancer 47:207–14.
  • Therasse P, Arbuck SG, Eisenhauer EA, et al. (2000). New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 92:205–16.
  • Eisenhauer EA, Therasse P, Bogaerts J, et al. (2009). New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 45:228–47.
  • Trotti A, Byhardt R, Stetz J, et al. (2000). Common toxicity criteria: version 2.0. An improved reference for grading the acute effects of cancer treatment: impact on radiotherapy. Int J Radiat Oncol Biol Phys 47:13–47.
  • Trotti A, Colevas AD, Setser A, et al. (2003). CTCAE v3.0: development of a comprehensive grading system for the adverse effects of cancer treatment. Semin Radiat Oncol 13:176–81.
  • Dindo D, Demartines N, Clavien PA. (2004). Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg 240:205–13.
  • Koopman M, Antonini NF, Douma J, et al. (2007). Sequential versus combination chemotherapy with capecitabine, irinotecan, and oxaliplatin in advanced colorectal cancer (CAIRO): a phase III randomised controlled trial. Lancet 370:135–42.
  • Lee DH, Oh SY, Lee YR, et al. (2011). A phase II study of modified FOLFOX4 for colorectal cancer patients with peritoneal carcinomatosis. Cancer Res Treat 43:225–30.
  • Hurwitz HI, Tebbutt NC, Kabbinavar F, et al. (2013). Efficacy and safety of bevacizumab in metastatic colorectal cancer: pooled analysis from seven randomized controlled trials. Oncologist 18:1004–12.
  • Overman MJ, Pozadzides J, Kopetz S, et al. (2010). Immunophenotype and molecular characterisation of adenocarcinoma of the small intestine. Br J Cancer 102:144–50.
  • Razenberg LG, van Gestel YR, Lemmens VE, et al. (2016). Bevacizumab in addition to palliative chemotherapy for patients with peritoneal carcinomatosis of colorectal origin: a nationwide population-based study. Clin Colorectal Cancer 15:e41–6.
  • Tamsma JT, Keizer HJ, Meinders AE. (2001). Pathogenesis of malignant ascites: Starling’s law of capillary hemodynamics revisited. Ann Oncol 12:1353–7.
  • Esquivel J, Piso P, Verwaal V, et al. (2014). American Society of Peritoneal Surface Malignancies opinion statement on defining expectations from cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in patients with colorectal cancer. J Surg Oncol 110:777–8.
  • Lam JY, McConnell YJ, Rivard JD, et al. (2015). Hyperthermic intraperitoneal chemotherapy + early postoperative intraperitoneal chemotherapy versus hyperthermic intraperitoneal chemotherapy alone: assessment of survival outcomes for colorectal and high-grade appendiceal peritoneal carcinomatosis. Am J Surg 210:424–30.
  • Elias D, Benizri E, di Pietrantonio D, et al. (2007). Comparison of two kinds of intraperitoneal chemotherapy following complete cytoreductive surgery of colorectal peritoneal carcinomatosis. Ann Surg Oncol 14:509–14.
  • McConnell YJ, Mack L, Francis WP, et al. (2013). HIPEC + EPIC versus HIPEC-alone: differences in major complications following cytoreduction surgery for peritoneal malignancy. J Surg Oncol 107:591–6.
  • Tan GH, Ong WS, Chia CS, et al. (2016). Does early post-operative intraperitoneal chemotherapy (EPIC) for patients treated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) make a difference?. Int J Hyperthermia 32:281–8.
  • Maillet M, Glehen O, Lambert J, et al. (2016). Early postoperative chemotherapy after complete cytoreduction and hyperthermic intraperitoneal chemotherapy for isolated peritoneal carcinomatosis of colon cancer: a multicenter study. Ann Surg Oncol 23:863–9.
  • Devilee RA, Simkens GA, van Oudheusden TR, et al. (2016). Increased survival of patients with synchronous colorectal peritoneal metastases receiving preoperative chemotherapy before cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Ann Surg Oncol 23:2841–8.
  • Ceelen W, van Nieuwenhove Y, Putte DV, Pattyn P. (2014). Neoadjuvant chemotherapy with bevacizumab may improve outcome after cytoreduction and hyperthermic intraperitoneal chemoperfusion (HIPEC) for colorectal carcinomatosis. Ann Surg Oncol 21:3023–8.
  • Eveno C, Passot G, Goéré D, et al. (2014). Bevacizumab doubles the early postoperative complication rate after cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (HIPEC) for peritoneal carcinomatosis of colorectal origin. Ann Surg Oncol 21:1792–800.
  • Cavaliere F, de Simone M, Virzi S, et al. (2011). Prognostic factors and oncologic outcome in 146 patients with colorectal peritoneal carcinomatosis treated with cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy: Italian multicenter study S.I.T.I.L.O. Eur J Surg Oncol 37:148–54.
  • Solass W, Kerb R, Mürdter T, et al. (2014). Intraperitoneal chemotherapy of peritoneal carcinomatosis using pressurized aerosol as an alternative to liquid solution: first evidence for efficacy. Ann Surg Oncol 21:553–9.
  • Nadiradze G, Giger-Pabst U, Zieren J, et al. (2016). Pressurized Intraperitoneal Aerosol Chemotherapy (PIPAC) with low-dose cisplatin and doxorubicin in gastric peritoneal metastases. J Gastrointest Surg 20:367–73.
  • Demtröder C, Solass W, Zieren J, et al. (2016). Pressurized intraperitoneal aerosol chemotherapy with oxaliplatin in colorectal peritoneal metastasis. Coloretal Dis 18:364–71.
  • Tempfer CB, Rezniczek GA, Ende P, et al. (2015). Pressurized intraperitoneal aerosol chemotherapy with cisplatin and doxorubicin in women with peritoneal carcinomatosis: a cohort study. Anticancer Res 35:6723–9.
  • Odendahl K, Solass W, Demtröder C, et al. (2015). Quality of life of patients with end-stage peritoneal metastasis treated with Pressurized Intraperitoneal Aerosol Chemotherapy (PIPAC). Eur J Surg Oncol 41:1379–85.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.