Review Open Access
Copyright ©The Author(s) 2019. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Feb 27, 2019; 11(2): 150-172
Published online Feb 27, 2019. doi: 10.4254/wjh.v11.i2.150
Colorectal liver metastases: An update on multidisciplinary approach
Felix Che-Lok Chow, Department of Surgery, Queen Mary Hospital, Hong Kong, China
Kenneth Siu-Ho Chok, Department of Surgery and State Key Laboratory for Liver Research, the University of Hong Kong, Hong Kong, China
ORCID number: Felix Che-Lok Chow (0000-0001-8800-9525); Kenneth Siu-Ho Chok (0000-0001-7921-3807).
Author contributions: Chok KSH conceptualized and supervised the study; Chow FCL did the literature review and wrote the manuscript; both authors approved the submitted version of manuscript.
Conflict-of-interest statement: None of the authors has any conflict of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Kenneth Siu-Ho Chok, FRCS(Ed), Associate Professor, Department of Surgery and State Key Laboratory for Liver Research, the University of Hong Kong, 102 Pok Fu Lam Road, Hong Kong, China. chok6275@hku.hk
Telephone: +852-22553025 Fax: +852-28165284
Received: October 5, 2018
Peer-review started: October 6, 2018
First decision: November 15, 2018
Revised: November 24, 2018
Accepted: December 4, 2018
Article in press: December 5, 2018
Published online: February 27, 2019

Abstract

Liver metastasis is the commonest form of distant metastasis in colorectal cancer. Selection criteria for surgery and liver-directed therapies have recently been extended. However, resectability remains poorly defined. Tumour biology is increasingly recognized as an important prognostic factor; hence molecular profiling has a growing role in risk stratification and management planning. Surgical resection is the only treatment modality for curative intent. The most appropriate surgical approach is yet to be established. The primary cancer and the hepatic metastasis can be removed simultaneously or in a two-step approach; these two strategies have comparable long-term outcomes. For patients with a limited future liver remnant, portal vein embolization, combined ablation and resection, and associating liver partition and portal vein ligation for staged hepatectomy have been advocated, and each has their pros and cons. The role of neoadjuvant and adjuvant chemotherapy is still debated. Targeted biological agents and loco-regional therapies (thermal ablation, intra-arterial chemo- or radio-embolization, and stereotactic radiotherapy) further improve the already favourable results. The recent debate about offering liver transplantation to highly selected patients needs validation from large clinical trials. Evidence-based protocols are missing, and therefore optimal management of hepatic metastasis should be personalized and determined by a multi-disciplinary team.

Key Words: Colorectal cancer, Liver metastases, Hepatic resection, Neoadjuvant therapy, Adjuvant chemotherapy, Intra-arterial therapy, Precision medicine, Multidisciplinary approach

Core tip: Surgery offers the only hope of cure in colorectal liver metastasis. It can be performed if complete metastasectomy is attainable. There is no consensus on the ideal management strategy for synchronous disease. A subset of patients presenting with unresectable disease may become eligible for resection after liver remnant augmentation or conversion therapy (chemo-therapeutics +/- biological agents). Amid increasing application of loco-regional therapies to colorectal liver metastasis, their role in the treatment paradigm remains to be defined. Refined patient selection – with greater emphasis on tumour biology – is essential to improving treatment outcome. The multidisciplinary approach helps determine the optimal treatment strategy from an expanding armamentarium of therapeutic options for each patient.



INTRODUCTION

Colorectal cancer (CRC) is a leading cause of tumour-related morbidity and mortality worldwide[1]. Approximately 50% of patients develop liver metastases (LM) in their course of disease[2]. Surgical resection is the only treatment that offers a chance of cure and long-term survival, with 5- and 10-year survival rates at around 40% and 25% respectively[3]. In selected cohorts, up to 97% of ten-year survivors remained disease-free after resection of colorectal LM (CRLM)[4]. However, only a minority of patients is suitable for upfront surgery. While improving surgical techniques and better adjuvant therapies are pushing forward the frontiers of resection, the importance of careful patient selection should not be overlooked. Not all patients undergoing resection enjoy long-term benefit – around 30% developed recurrence and 15% succumbed to their disease within a year after surgery[5]. A personalized treatment approach – taking tumour biology, disease staging and patient condition into consideration – is the key to improving outcomes.

Management of stage IV CRC is optimized by grouping relevant specialists together under a multidisciplinary team (MDT) setting[6]. A proficient MDT consists of at least a colorectal surgeon, a liver surgeon, medical and radiation oncologists, a radiologist, a pathologist and a case nurse[7]. Better patient and disease evaluation, joint decision-making and optimization of multimodal therapy not only improve patient outcome, but also enhance consistency and coordination of care. The value of multidisciplinary approach in the management of gastrointestinal malignancies has been demonstrated in a prospective study. Despite 84% of clinicians being certain of their original plan before discussion, a change was recommended in 36% of cases, 72% of which were major[8]. This review highlights current controversies and relevant evidence in the management of CRLM.

PATIENT AND TUMOUR EVALUATION

Accurate assessment of a patient’s general health condition (comorbidities, performance status and liver function) and the extent of disease is important for treatment planning.

Radiological assessment

Guidelines recommend CT scan of thorax, abdomen and pelvis for initial workup. It is adequate for determining resectability in most cases. In cases of doubt, a second imaging modality such as MRI could be added[9,10]. In patients planning for upfront resection, sensitivity of MRI and CT were similar (94% vs 91%)[11]. However, in general, MRI was more sensitive than CT in detecting CRLM (91% vs 82%), particularly for sub-centimetre lesions or reassessment after neoadjuvant chemotherapy (when the sensitivity of CT dropped to 77%)[12]. Gadoxetic contrast further increased the diagnostic confidence of MRI to 98.3%, compared with 85.7% and 65.2% in conventional contrast MRI and CT[13]. High-quality baseline imaging is essential before any chemotherapy, when lesions are more readily detectable; whereas comparison with post-chemotherapy films gauges treatment response and delineates tumour biology. MRI is useful when characterization is difficult e.g., underlying fatty liver, or multiple small nodules with uncertain nature.

Controversy 1: Role of positron emission tomography - computed tomography: Whether positron emission tomography - computed tomography (PET-CT) offers additional information to CT and/ or magnetic resonance imaging (MRI) is controversial. An early randomized study showed PET (without CT) significantly reduced the number of futile operations (28% vs 45% in control) and prevented an unnecessary surgery in every 6 patients[14]. In another randomized trial, however, PET-CT did not influence decision-making in patients with resectable CRLM – the PET-CT group had similar hepatic resection rate and survival as the controls; it only altered surgical management in 8% patients (2.7% did not undergo surgery and 3.4% underwent additional organ surgery)[15]. Long-term follow-up of this trial concluded PET-CT did not improve disease-free or overall survival[16]. According to a meta-analysis, PET-CT was less sensitive but more specific than CT or MRI in detecting CRLM – sensitivity 66% vs 79% vs 89%; specificity 86% vs 67 vs 81%[17]. In our practice, we still perform PET-CT in the majority of patients to assess for extra-hepatic disease (EHD). As an adjunct to CT, its value in staging EHD was evident in 20% patients – preventing futile operations, guiding resection of loco-regional nodal disease, or clarifying indeterminate CT findings[18].

Evaluation of future liver remnant (before major hepatectomy)

Accurate preoperative estimation of liver functional reserve is essential to prevent post-hepatectomy liver failure, especially in patients with extensive tumour load or highly compromised livers. CT volumetry is routinely used before major hepatectomy. In most centres, future liver remnant (FLR) volumes of 25% and 40% are accepted as adequate for normal and diseased liver respectively. However, FLR volume does not necessarily reflect its function, particularly as quality of liver tissue can be affected by pre-operative chemotherapy[19].

Indocyanine green (ICG) clearance is a long-established functional test for selecting surgical candidates with adequate liver reserve; its determination by pulse dye densitometry and intra-operative application have attracted great interest[20,21]. The use of ICG has limitations though: its uptake by hepatocytes can be impaired by hyperbilirubinemia, and it reflects the total liver function rather than specifically the FLR performance, failing to address regional variations within the liver[19]. Segmental hepatic function can be measured by hepatobiliary scintigraphy; the commonest used agent being Technetium-99m (99mTc) labelled mebrofenin, which is taken up by hepatocytes and directly excreted into the biliary tree. Using a single cut-off value of 2.7%/min·m2 irrespective of the liver tissue quality, 99mTc-mebrofenin hepatobiliary scintigraphy has been shown to outperform CT volumetry in predicting the risk of post-hepatectomy liver failure[22]. Nonetheless, further evidence is required to support its widespread use in clinical practice.

PATIENT SELECTION AND PROGNOSTIC PREDICTION

Although hepatic resection gives the best results on a population level, not everyone with technically operable disease benefits from surgery[5]. Appropriate patient selection ensures the offered intervention, surgical or systemic, is optimal to each particular patient.

Resectability of a particular CRLM should be determined in a multidisciplinary setting, with input from hepatobiliary surgeons, oncologists, radiologists and pathologists. Apart from pure technical considerations, there is a growing emphasis placed on oncological resectability[23]. The former focuses on whether a margin-negative (R0) resection can be achieved while preserving a liver remnant comprised of two contiguous segments with adequate volume, function, vascular inflow and outflow, as well as biliary drainage. Oncological/prognostic evaluation aims to select patients with higher likelihood of cure or sustained disease remission; taking tumour biology (in particular disease progression/remission during neoadjuvant therapy), mutation status, intra-hepatic tumour burden and extent of EHD into consideration. Resection criteria based on the number, maximal size and distribution of tumours no longer apply; instead resectability should be defined case-by-case based on different prognostic factors. With continued advancement in surgical technology, systemic therapies and multimodality treatment, the definition of resectability will continue to evolve and expand to cover advanced diseases once deemed non-resectable.

Traditional clinico-pathological prognostic factors include[24-26]: (1) Characteristics of primary CRC, e.g., advanced T stage, nodal status, location of tumour (right sided tumour associated with poorer outcome); (2) Factors related to CRLM e.g., size of largest liver metastasis, number of lesions, grade of differentiation, margin status; (3) Presence of extrahepatic disease; (4) Elevated serum carcinoembryonic antigen (CEA) level; and (5) Disease-free interval between primary CRC and metachronous CRLM

In clinical practice, the predictive value of each individual factor is limited. Several scoring systems were devised aiming to provide an overall risk assessment; the most widely quoted one is the Fong clinical risk score (assigning points to positive margin, EHD, node-positive primary, disease-free interval from primary to CRLM, more than one solitary LM, largest LM > 5 cm, and CEA > 200 ng/mL)[27]. These systems, however, failed to demonstrate predictive accuracy for long-term survival or in the neoadjuvant setting across institutions[28,29]. Their clinical utility remained uncertain.

Both radiological and pathological response to preoperative chemotherapy predict better survival after resection of CRLM[30,31]. On CT or MRI scans, treatment response can be judged by degree of tumour shrinkage and morphological changes unrelated to size (e.g., tumour density, tumour-liver interface). Due to the limitations of the Response Evaluation Criteria in Solid Tumours (RECIST), new parameters like early tumour shrinkage and depth of response have been proposed to aid in prognostication[32,33]. 18-fluorodeoxyglucose (18FDG) PET/CT also has a role in prognostication–LM with high glucose metabolism [high pre-treatment standardized uptake value (SUV)] and poor metabolic response to systemic therapy had poorer overall and disease-free survivals[34]. Complete pathological response, on the other hand, has been associated with an excellent 5-year survival of 76%[35].

All these clinical, radiological and pathological characteristics are considered surrogate markers of the underlying tumour biology. There is growing interest in directly assessing tumour biology by molecular profiling and integrating biomarkers into prognostication systems. KRAS gene has been extensively studied. As there was a high concordance of KRAS status between primary CRC and CRLM, it can be evaluated on biopsy or resected specimens of the primary tumour[36]. Apart from predicting poor response to anti-epidermal growth factor receptor (EGFR) therapies, KRAS mutation has been associated with higher rates of EHD, adverse response to chemotherapy, positive resection margin, worse overall and recurrence-free survivals after surgery irrespective of the chemotherapy regimen, and poorer survival after re-resection for recurrence[25,37,38]. In view of the poor prognosis, some suggested aggressive treatment might not be worthwhile for KRAS mutant tumours with multiple risk factors (e.g., node-positive primary, individual CRLM > 3 cm, more than 7 cycles of systemic chemotherapy given)[39]. Incorporating KRAS mutation status to traditional risk scores might improve survival prediction[40]. BRAF, another commonly tested gene in CRC, also predicted survival outcome in CRLM. BRAF mutated tumours were refractory to standard chemotherapy and anti-EGFR agents and had far inferior survival; resection should only be offered to those with limited disease, taking note of the high risk of recurrence[41,42]. By using gene expression microarray on resected CRLM, a 20-gene molecular risk score was externally validated to be an independent prognosticator of overall and recurrence-free survival after liver metastasectomy; it was more accurate than traditional clinical risk scores[43]. With further research, risk stratification and individualized therapy based on molecular profiling could be realized in the foreseeable future.

Nowadays we realize tumour heterogeneity exists not only amongst different patients, but also within individual tumours and among metastatic sites. Multiple cancer subclones coexist and evolve simultaneously, with treatment acting as selection pressure. Tumour biopsy at a single site at a particular time cannot reflect the entire disease throughout the treatment period[25]. Serial imaging has the advantage of assessing multiple tumour locations in a longitudinal fashion, but is frequently limited by spatial resolution. The role of circulating liquid biomarkers is under investigation; its collection at several time points (e.g., pre-treatment, after each cycle of chemotherapy, before and after resection) can offer insight into the evolving tumour biology. Presence of circulating tumour cells (CTCs) predicted impaired survival in CRLM; it could also be a source to detect KRAS and BRAF mutations to guide choice of targeted therapy[44,45]. Plasma level of circulating cell-free DNA (cfDNA) has been proven a predictor of survival in metastatic CRC[46]. Early work showed cfDNA sequencing allowed identification of gene mutation or micro-satellite instability[47]. Its detection and analysis may improve diagnostic efficiency in both screening and surveillance settings[48]. MicroRNA provides a molecular snapshot of intracellular activity within cancer; its signature has been shown to predict metastasis and prognosis in CRC[49]. If microRNA profiling can be obtained from serum samples, tumour prognostication from blood tests is distinctly feasible.

NEOADJUVANT TREATMENT
Controversy 2: Role of neoadjuvant therapy in clearly R0 resectable CRLM

Theoretically, neoadjuvant therapy allows assessment of the natural history of disease before embarking on metastasectomy. It potentially shrinks the tumour and reduces the extent of liver resection, treats micro-metastases thereby lowering recurrence rate, as well as guides further therapeutic plan based on disease response to treatment. Its benefit is not proven from an evidenced-based point of view though.

Earlier studies confirmed an objective radiological response could be expected in two-thirds of treated patients[50]. However, the majority of retrospective studies failed to demonstrate any overall survival (OS) benefits from neoadjuvant therapy – five-year OS rates ranged from 38.9% to 74% in patients who had pre-operative chemotherapy before liver resection, compared with 20.7 to 56% in patients who underwent upfront surgery[51]. A landmark randomized controlled trial (the EORTC intergroup trial 40983) showed perioperative FOLFOX (folinic acid, fluorouracil, and oxaliplatin; 6 cycles before and 6 cycles after surgery) improved 3-year progression-free survival (PFS) modestly – 42.4% compared with 33.2% in surgery-only patients, an absolute 9.2% increase – at the cost of higher peri-operative morbidity (25% vs 16%). This did not translate into any overall survival benefit at a median follow-up of 8.5 years[52,53]. A meta-analysis including 18 studies concurred neoadjuvant treatment, in general, did not offer PFS or OS advantage; however, it could improve survival in patients considered high risk of recurrence (pooled hazard ratio for 5-year OS = 0.69)[54]. The CHARISMA randomized trial is underway to investigate whether neoadjuvant XELOX improves survival in high-risk CRLM patients[55].

Based on current evidence, the European Society for Medical Oncology (ESMO) guidelines suggested the need for perioperative systemic therapy is defined by “technical criteria for resection and prognostic considerations”. Upfront surgery is justified in patients with clearly resectable disease and favourable prognosis; while peri-operative FOLFOX or XELOX should be considered when resectability or prognostic criteria is unclear or “not excellent”[9]. In our centre, we favour the surgery-first approach as long as R0 resection can be attained, because the unclear survival benefit of neoadjuvant treatment carries with it the risk of chemotherapy-associated liver injury (discussed below). Future research should focus on accurately defining “high-risk” patients who will benefit most from preoperative therapy.

Regimens and potential risks

Current guidelines suggest oxaliplatin-based doublet chemotherapy (FOLFOX/ XELOX) as the neoadjuvant treatment-of-choice for resectable CRLM, while FOLFIRI or FOLFOXIRI are alternatives[9,10]. A meta-analysis showed the addition of molecular targeted therapy conferred a higher overall response rate than chemotherapy alone (68% vs 43%), but did not improve survival[56]. A lack of PFS benefit was also observed in the New EPOC trial, which studied the effect of combining EGFR-inhibitor (cetuximab) to perioperative systemic chemotherapy; patients who received cetuximab actually experienced worse PFS (14.1 vs 20.5 months in control)[57]. Given these results, cetuximab should not be added to standard perioperative chemotherapy regimens. Bevacizumab, an anti-vascular endothelial growth factor (VEGF), plus FOLFIRI in the neoadjuvant setting yielded a response rate of 66.7% in resectable CRLMs[58]; whether this translates into any survival benefit remains to be investigated. The ongoing PERIMAX trial compares perioperative bevacizumab plus FOLFOXIRI with adjuvant FOLFOX; with the primary endpoint being failure-free survival[59].

The potential risks of perioperative chemotherapy include disease progression during treatment and hepatotoxicity. Initially resectable CRLM may progress despite using the best available chemotherapeutics, and become unresectable or require a more extensive surgical approach; the rate was 7% in the EORTC trial[52]. From another perspective, this small group of patients have very aggressive tumour and their disease would progress despite any treatment given; some argue neoadjuvant treatment only selects them out and prevents futile operations.

Chemotherapy-associated liver injury (CALI) can occur with commonly used regimens. Oxaliplatin was associated with sinusoidal obstruction syndrome in up to 38% patients, while steatosis and steatohepatitis complicated 9.3% patients receiving irinotecan[60]. Patients with severe sinusoidal dilation (OR 1.73) or steatohepatitis (OR 2.08) were more likely to suffer from postoperative major morbidity and liver surgery-specific complications[61]. It has been shown postoperative complication rate increases if the interval between chemotherapy completion and surgery is too short (< 4 wk) or too many cycles (>9) of preoperative chemotherapy are given[62,63]; limiting the duration of neoadjuvant therapy and ensuring recovery of liver function before operation can reduce the impact of CALI. Future research should focus on identifying patients at risk of CALI and devising liver protective strategies for such patients.

Disappearing LM

A subset of CRLMs can totally vanish on imaging after neoadjuvant treatment; they were referred as disappearing LMs (DLMs). This phenomenon is related to the quality of imaging, particularly as chemotherapy can compromise radiological detection of CRLM[64]. The key question is whether the lesions have been truly cured or just being missed by suboptimal post-neoadjuvant scans.

MRI has the highest sensitivity and is the preferred imaging modality in this setting[65]. In existing literature, the percentage of patients with one or more DLMs ranged widely from 7% to 37%; however, this phenomenon may be over-reported as most studies only utilized CT and intraoperative ultrasound for reassessment[66]. Complete disappearance of all initial CRLMs is rare, with an incidence of 0%-6%; therefore, most patients still undergo surgery. In laparotomy, macroscopic residual disease could be detected at 11%-67% DLM areas, highlighting the inadequacy of current post-neoadjuvant imaging. Importantly, there was clear difference between complete radiological and pathological responses – when areas of DLM were resected, microscopic residual disease was found in up to 80% of specimens[67].

Only surgery potentially offers cure; the phenomenon of DLM should be prevented, albeit complete pathological response has been associated with good prognosis[35]. To reduce the risks of DLMs, preoperative over-treatment has to be avoided – some proposed restaging after every 3 cycles of neoadjuvant therapy to enable earlier decision of surgery, and limiting the entire course of chemotherapy to 4 to 6 cycles[66,68]. Using a combination of imaging modalities enhances the detection of residual metastases and lowers the incidence of false DLMs. As conservative management i.e. leaving DLMs in-situ resulted in a local recurrence rate of 19% to 74%[66], DLMs should be resected whenever feasible. Some proposed even in patients with complete radiological resolution of all CRLMs, surgical exploration may be warranted for meticulous intra-operative assessment. Whether resection offers survival benefit is unclear though; patients with untreated DLMs could have comparable OS to those who underwent complete surgical treatment, in spite of a higher intrahepatic recurrence rate[69]. In selected patients, leaving certain DLMs untreated may be reasonable but this decision should only be made in a multi-disciplinary setting[23,68,70]. Ablation of DLM sites is an appealing alternative, but there is no evidence proving its efficacy to date. Initial experience with computer-guided resection of DLMs showed promising results. Augmented reality is a technology fusing reconstructed pre-treatment CT images with real-time patient images, thereby facilitates DLM localization and ensures clear resection[71].

SYNCHRONOUS DISEASE

About one sixth of CRC patients had LM at presentation[72]. Defined as CRLM detected concurrently or before the primary CRC, synchronous disease has been shown to have less favourable cancer biology and post-resection survival – 5-year survival was 39%, compared with 48% in metachronous CRLM[7]. Western guidelines and expert consensus recommend neoadjuvant chemotherapy in this higher-risk setting, unless resection at both sites are considered easy[7,9]. However, an analysis based on the Liver Met Survey International Registry showed neoadjuvant therapy offered no survival advantage in resectable synchronous CRLM, with a 5-year OS of 42% similar to 47% in the upfront surgery group[73]. In Asian countries, preoperative chemotherapy is not a standard in resectable synchronous disease.

Controversy 3: Optimal surgical sequencing for resectable synchronous disease

To date, there has been no randomized trial on the surgical approach to synchronous disease. The optimal strategy remains controversial. Decision should be individualized taking into consideration of patient’s fitness and tumour status – whether the primary CRC is symptomatic or obstructive (CRC needs to be resected first under these circumstances), as well as the extent of CRLM and magnitude of liver resection required. Treatment strategy is best determined in multi-disciplinary setting, which has been associated with greater likelihood of simultaneous resection[74].

The conventional approach comprises of resection of the primary CRC followed by liver metastasectomy, commonly with chemotherapy applied between the two surgeries. This avoids potential complications of the primary tumour, but carries significant risk of CRLM progression beyond resectability; less than 30% patients completed the whole treatment and underwent liver resection[75].

The liver-first approach, also known as the reversed strategy, may be more appropriate if an asymptomatic colon primary coexists with extensive CRLM or the primary tumour is a locally advanced rectal cancer (which needs neoadjuvant chemoirradiation). After systemic chemotherapy, patients underwent liver metastasectomy prior to removal of primary CRC. Given hepatic metastases rather than the primary tumour dictates these patients’ prognosis, early administration of chemotherapy to control liver and systemic diseases optimizes the chance of potentially curative hepatic resection and long-standing survival[76]. Compared to the classical approach, more patients (around three quarters) could complete the whole paradigm and underwent both liver and colorectal resection[77]. In spite of a respectable 5-year OS of 33.1% and acceptable perioperative morbidity and mortality rates of 31.5% and 3.3%, a recent single-centre study reported a high recurrence rate of 51.4% after a median follow-up of 20.9 mo[78]. Whether liver-first approach confers survival benefit remains undetermined.

Combined colorectal and liver resection in one single operation is generally reserved for patients with easy-to-resect primary tumours and limited hepatic disease[7]. Traditionally this approach was associated with increased morbidity, including infectious liver and anastomotic complications. With technological advancement, outcomes of combined resection have improved. Provided resection at the other site is minor, incorporating a major liver or rectal resection into this approach is deemed safe[79,80]. Cumulative morbidity and mortality rates were comparable to, or even better than, staged procedures; long-term oncological outcomes were similar – 1- and 5-year OS rates for one-stage operation were 90.5% and 38.5% respectively, while 5-year DFS was 25.3%[81,82]. A recent meta-analysis of 30 studies confirmed simultaneous liver and colorectal resection was associated with shorter hospital stay compared with staged procedure, without adversely affecting perioperative morbidity or long-term survival[83]. This approach is gaining popularity as more evidence proved its safety and efficacy.

A meta-analysis comparing the classical, synchronous and liver-first approaches did not show any difference in surgical outcomes or survival advantage[84]. Because of the limited evidence available, the optimal treatment strategy is unclear. Choice of procedure should be personalized and based on expertise available in different institutions.

In the presence of unresectable metastases, the benefit of resecting an asymptomatic primary without liver surgery is debatable. A meta-analysis showed primary tumour resection conferred a survival benefit of 6.4 months compared with chemotherapy alone, however the result is questionable given the significant selection bias[85]. To date, no randomized trial was completed to clarify this issue.

HEPATIC RESECTION

CRLM resection offered an overall median survival of 3.6 years; five- and 10-year survival ranged from 16% to 74% (median 38%) and 9% to 69% (median 26%) respectively[3]. Following major hepatectomy for CRLM, large contemporary series reported perioperative mortality and major morbidity rates of 1%-5% and approximately 20% respectively.

Surgical approach

Despite the addition of systemic chemotherapy, intra-hepatic recurrence is common after surgery of curative intent and occurred in two-thirds of patients within 3 years[5]. Increasing evidence showed re-resection of liver-limited recurrence is safe and can produce good long-term outcomes in selected patients[86]. Compared with anatomical resection, parenchymal-sparing hepatectomy (PSH) enhances the likelihood of repeated resection in the event of intra-hepatic recurrence. Both approaches have equivalent safety profile and oncological outcomes (R0 resection rate, liver recurrence-free survival and OS) after the index operation. However, in case of recurrence, PSH has been associated with a better 5-year survival since more patients could underwent salvage surgery[87,88]. Nowadays, PSH is considered the standard approach to CRLM unless it is precluded by the anatomy of the disease.

Amid growing interest in laparoscopic liver resection (LLR), the recently published OSLO-COMET randomized controlled trial demonstrated the safety and short-term efficacy of laparoscopic minor PSH for CRLM. Compared with the open-surgery group, the LLR group had less postoperative complications and a shorter hospital stay, while 90-d mortality and percentage of involved resection margins were similar[89]. For oncological outcomes, both approaches had comparable tumour recurrence rate, DFS, and 5-year OS in a meta-analysis of propensity-score matched studies; the LLR group even had better 3-year OS[90]. While there were evidence supporting the use of minimally invasive techniques for minor CRLM resection, its role in major liver surgery and challenging tumour locations needs further clarification. The advanced laparoscopic skills required can limit its widespread application. There are ongoing efforts to facilitate the education and implementation of LLR worldwide[91].

Width of resection margin

Contrary to the historic “1-cm rule”, several large series showed as long as R0 resection (≥ 1 mm) is achieved, extra margin width does not add DFS/OS advantage[92-94]. From observations that (1) survivals were similar between R0 and R1 resections if tumour showed optimal radiological or pathological response to neoadjuvant chemotherapy[95]; (2) most recurrences occurred outside the surgical margin in a disseminated manner; and (3) margin re-resection from R1 to R0 did not improve long-term outcomes[96], positive margin status could just be a surrogate of aggressive tumour biology. The latter is increasingly recognized as the most important prognostic determinant. While an expected close margin (especially when tumour is close to vital vessels) is not a contraindication to surgery, surgeons should aim at wider margins to ensure a R0 resection[97].

Controversy 4: Role of surgery in the presence of extrahepatic disease: Management of CRLM in this setting remains controversial. Albeit defined as a poor prognostic factor, limited extrahepatic disease (EHD) does not contraindicate liver surgery; the prerequisite is all diseases including the primary CRC, LM and EHD can be completely resected or controlled[9,98]. Resection of CRLM together with concurrent EHD significantly improved the 5-year OS from 0% to 28%, with a median survival of 31 mo[99].

The location of EHD matters – lung metastasis carries a more favourable prognosis than peritoneal or portal/para-aortic nodal metastases (5-year OS being 26%, 17% and 15% respectively after complete resection of CRLM and EHD)[100]. According to data from the LiverMetSurgery registry, resection of concurrent liver and lung metastases was associated with similar OS as isolated liver metastasectomy[101]. The relative indolent nature of most colorectal pulmonary metastases supports CRLM resection in the presence of unresectable lung disease – 5-year OS of 13.1% was better than 1.6% achieved by chemotherapy alone, but as expected much worse than the 56.9% after complete metastasectomy at both sites[102].

Although long-term survival is possible, true definitive cure is rare after resecting concurrent CRLM and EHD. In a retrospective review, disease recurred in 90.2% patients at a median of 8 mo, 85% being systemic recurrence; 5-year recurrence-free survival was 5%[103]. Effort was made to identify subgroups who benefit most from radical surgery. For example, in patients with synchronous LM and peritoneal carcinomatosis (PC) of colorectal origin, complete cytoreductive surgery and liver metastasectomy followed by intra-peritoneal chemotherapy achieved a median OS of 40 mo in those with limited PC (peritoneal cancer index, PCI < 12) and 1-2 LM. Outside these criteria, the survival benefit of radical surgery was significantly reduced – patients with PCI ≥ 12 or at least 3 LM had a median OS of 27 mo[104]. Non-pulmonary EHD, EHD concomitant to LM recurrence, CEA ≥ 10 ng/mL, more than 5 LM and right-sided colonic primary all predict poor prognosis after surgery for concurrent CRLM and EHD; no patients with more than 3 factors achieved 5-year survival[105]. However, this prognostication system has not been externally validated.

In summary, complete resection of all concurrent metastases can improve disease control and survival in selected patients, although cure is rare and recurrence is expected. Future research can help develop prognostic scores to better select patients for radical surgery.

Controversy 5 – Ablation or surgical resection? Thermal ablation involves destruction of cancer by heat, with radiofrequency ablation (RFA) and microwave ablation (MWA) being the most widely employed and studied modalities. Despite showing advantage of fewer complications and better post-procedural quality of life, RFA was inferior to hepatic resection in terms of survival (lower OS and DFS) and recurrence (higher rates of local, intra-hepatic and any recurrences)[106-108]. Surgical resection is the choice of treatment in young, fit patients, based on its proven long-term efficacy and lower recurrence rate.

In small tumours less than 3 cm, however, RFA may attain comparable oncological outcomes to resection. Following ablation to complete tumour necrosis with a margin over 5 mm, one-year local disease progression rate as low as 3% has been reported[109,110]. In high-risk patients (elderly, or with significant comorbidities), the considerably lower morbidity associated with ablation could justify its use over resection, provided patients accept the trade-off of potentially inferior long-term results. The ongoing LAVA randomized trial is designed to clarify the optimal treatment strategy in these high-risk individuals[111].

Borderline resectable disease due to insufficient liver remnant: Patients with extensive bilobar LM poses a unique challenge. The major limiting factor for a curative resection is the volume and function of the future liver remnant (FLR). Different management strategies and their advantages and disadvantages were listed below.

Portal vein embolization (PVE) is a well-established method for augmentation of small liver remnant, thereby enables extensive curative resection initially deemed high risk of postoperative liver failure. By obliterating portal flow to the liver segments involved by metastases (intended to be resected) and diverting it to the side that will remain, PVE is expected to increase the FLR volume by 43.1% on average. Less than 5% patients had inadequate hypertrophy response precluding them from the planned resection[112]. In one study, extended right hepatectomy became feasible after PVE in two thirds of patients who initially had inadequate FLR, yielding a survival similar to those who did not need PVE[113]. There were concerns about post-PVE tumour progression leading to unrectable disease, which could occur in up to 20-30% patients; whether PVE itself accelerates tumour growth remained controversial. Continued administration of chemotherapy after PVE has been shown to reduce tumour progression rate, while minimizing the interval between PVE and resection to 4 wk has also been advocated[114]. Reassuringly a meta-analysis showed PVE did not adversely affect overall survival or intrahepatic recurrence in patients undergoing major liver resection for CRLM[115]. Future research goals include identification of individuals at risk of rapid tumour progression and strategies to reduce progression rate without compromising FLR hypertrophy.

Two-stage hepatectomy (TSH) aims to remove all CRLM in two sequential operations for selected patients with advanced bilobar disease, in whom removing all lesions with safe margin is impossible during a single procedure. Pre-operative chemotherapy was frequently employed to select patients with favourable tumour biology. Following the first stage (for tumour clearance of FLR), a 2- to 3-mo interval allows liver tissue to regenerate (often aided by PVE) before a second stage hepatectomy took place to achieve R0 resection. A systematic review reported three quarters of patients successfully underwent second stage hepatectomy and R0 resection rate was 75%; the main reason for non-completion was interval disease progression (88%). Postoperative morbidity rate was 17% and 40% after first and second stage respectively, and overall mortality was less than 5%. A median OS of 37 mo and a 3-year DFS rate of 20% were encouraging, and comparable to other series of lesser-scale CRLM resections[116].

Both PVE and TSH, although proven effective, have a considerable patient drop-out rate due to tumour progression in the waiting interval. A novel surgical technique was introduced in 2012 to treat advanced bilobar liver tumour in a more rapid fashion.

Associating liver partition and portal vein ligation for staged hepatectomy (ALPPS) comprises liver transection and ligation of portal vein in the first operation, followed by resection of the diseased liver segment 7-14 d later. By redistributing total portal blood flow to FLR and triggering an inflammatory response, ALPPS induces rapid hypertrophy of FLR at an estimated growth rate of 22-35 mL/d (compared with 3-5 ml/d after PVE)[117]. The benefits of ALPPS include a greater FLR hypertrophy (76% vs 37% for PVE) and a higher rate of completion of stage 2 hepatectomy (100% vs 77% for PVE)[118]. A recent randomized controlled trial confirmed ALPPS had superior resection rate compared with TSH in patients with advanced CRLM, 92% vs 57%[119]. ALPPS has also been reported as a salvage treatment for failed PVE[120]. However, these came at the price of higher morbidity and mortality. The relative risks for overall and major morbidity (e.g., bile leakage and sepsis) were 1.39 and 1.57 compared with TSH[117]. As per the international ALPPS Registry, the 90-d mortality rate was high at 8.8%, three quarters were due to postoperative liver failure[121]. Oncological outcomes after ALPPS were also unclear; limited data available came from case series only. For CRLM, recurrence rates ranged from 14.3% (after a median follow-up of 9 mo) to 78.3% (after a median of 22.5 mo)[122,123]. Patients with “otherwise unresectable” advanced CRLM were identified from the international ALPPS registry, overall survival after ALPPS was not superior to matched controls who received palliative systemic treatment (with median OS of 24.0 mo vs 17.6 mo, P = 0.88)[124]. To improve outcomes of this radical surgical strategy, careful patient selection is essential; a consensus on the ideal indications of ALPPS is urgently needed. Recent promising results from high-volume centres suggest that, with surgical advancement, ALPPS can have low perioperative risk (0% mortality and 21% severe complications) and satisfactory survival (3-year OS 50% and 3-year DFS 13%) in experienced hands[125]. However, given the unclear long-term outcome and questionable safety profile, currently ALPPS should be limited to high-volume institutes in research setting.

Combining ablation with resection allows potential tumour clearance in extensive bilobar CRLM. This approach could achieve long-term survivals (5-year OS 37%-56%) comparable to that of TSH, with improved perioperative outcomes – less blood loss and shorter length of stay. The short disease-free interval (median DFS around 9 mo), though, suggested a temporary disease control rather than complete cure[126-128].

ADJUVANT THERAPY (AFTER POTENTIALLY CURATIVE METASTASECTOMY)

Adjuvant systemic therapy aims to reduce recurrence and prolong survival after curative resection, ideally with minimal treatment-related toxicity. In primary CRC, postoperative oxaliplatin-containing chemotherapy has been shown to offer survival benefit for up to 10 years after curative resection for stage III disease[129].

The role of adjuvant treatment is more controversial for CRLM; according to existing evidence, it only improves DFS but not OS. From pooled analysis of two early randomized trials’ results (French FFCD trial and English ENG trial), adjuvant fluorouracil potentially improved OS (median 62.2 mo vs 47.3 mo, P = 0.095) and PFS (median 27.9 mo vs 18.8 mo, P = 0.058) compared with surgery alone, but both did not reach statistical significance[130]. Adjuvant oral uracil-tegafur and leucovorin also only prolonged recurrence-free survival (from 0.70 to 1.45 years) without improving OS, as demonstrated by a Japanese RCT[131]. Current recommendations for oxaliplatin-containing adjuvant regimen (FOLFOX) were based on extrapolation of result of the EORTC intergroup trial 40983, which showed perioperative FOLFOX conferred a PFS benefit but did not affect OS[53]. The JCOG0603 trial is now underway to determine if adjuvant mFOLFOX is superior to resection alone[132]. Meanwhile, adjuvant FOLFIRI did not improve DFS or OS compared with 5FU alone, but was associated with lower treatment tolerance in a randomized study[133]; therefore, FOLFIRI is not commonly used in the adjuvant setting for CRLM. According to the latest ESMO guidelines, there was no strong evidence supporting the use of adjuvant chemotherapy in patients with good oncological and technical criteria who underwent upfront surgery; on the other hand, patients with unfavourable prognostic criteria or have not received any previous chemotherapy for metastatic disease may benefit from adjuvant therapy (e.g., FOLFOX/ XELOX)[9].

To date, no evidence supports the combined use of chemotherapeutics and biological targeted agents in the adjuvant setting after resection of CRLM. Addition of bevacizumab to modern chemotherapy or combination of hepatic arterial chemotherapy infusion (HAI) and systemic chemotherapy did not prolong survival, but appeared to increase biliary toxicity[134,135]. A randomized trial is in progress to assess whether bevacizumab gives additional benefit to adjuvant XELOX[136]. As the New EPOC trial linked its use in the peri-operative setting to a shorter PFS, cetuximab is generally not recommended in the adjuvant setting after liver metastasectomy[57].

Taking advantage of CRLM’s predilection of hepatic artery neovascularization, infusion of cytotoxics via the hepatic artery can deliver high concentration of therapeutic agents to the tumour while minimizing side-effects. An earlier RCT showed adding floxuridine-HAI to systemic fluorouracil (5-FU) chemotherapy improved PFS but not OS after a median follow-up of 10 years[137]. In a recent retrospective study, adjuvant HAI offered an OS advantage of approximately 2 years compared to systemic chemotherapy alone, and this benefit was substantiated in patients receiving modern chemotherapy[138]; the 5- and 10-year OS reached 78% and 61%[139]. The ongoing PACHA-01 trial compares the outcomes of adding oxaliplatin-HAI vs systemic oxaliplatin to adjuvant systemic 5-FU after resection or thermal ablation of at least four CRLM[140]. More evidence is needed to show whether HAI offers additional benefit to modern doublet or triplet adjuvant chemotherapy. However, the unique expertise required and the need of placing a special port-catheter has limited its use in specialized centres only.

UNRESECTABLE LIVER-ONLY OR LIVER-DOMINANT METASTASES – A MULTIDISCIPLINARY APPROACH

As mentioned, resectability should be determined by a MDT on a per patient basis, taking into consideration of technical and oncological factors. For unresectable CRLM, the former standard of care is palliative systemic chemotherapy. Although modern doublet or triplet chemotherapy (FOLFOX, FOLFIRI, FOLFOXIRI) have considerably improved the median OS to 15-21 mo and these were further extended by targeted agents, long-term survival remained rare[141,142]. In an attempt to obtain durable local disease control or even cure, two approaches have been employed – (1) conversion therapy followed by potential curative resection; and (2) use of single or a combination of liver-directed therapies. For systemic treatment regimen and local therapy, clinicians can determine the most appropriate strategy from a ‘toolbox’ of ever-expanding options, according to patient and disease factors, treatment goal and its related morbidity[9]; again, the decision is best made in a multi-disciplinary setting.

CONVERSION CHEMOTHERAPY

A subset of patients with initially unresectable CRLM (around 15%-30% depending on the definition of unresectability) may be rendered resectable after conversion chemo-therapy. In a systematic review of 10 studies using different downsizing regimens, an objective radiological response was achieved in 64% (range 43%-79%) patients; 22.6% underwent macroscopically curative liver resection (most studies reported a range of 12.5%-45%) and R0 resection rate was 87%. The median OS and DFS after liver metastasectomy were 45 and 14 months respectively[143].

The optimal regimen for conversion to operable disease remains unclear. Standard doublet chemotherapy FOLFOX or FOLFIRI had conversion rates between 9% to 33%[144]. Compared with FOLFIRI, intensified triplet chemotherapy FOLFOXIRI improved the secondary R0 resection rate from 12% to 36%, median PFS from 6.9 to 9.8 mo, and median OS from 16.7 to 22.6 mo; albeit at the cost of greater but manageable toxicity e.g., peripheral neuropathy and neutropenia[145]. Addition of targeted agents is recommended by guidelines, but there is no concrete supporting evidence. In a large RCT, giving bevacizumab together with XELOX/ FOLFOX only moderately improved resectability (from 6.1% to 8.4%) and PFS (from 8 to 9.4 mo), but did not prolong OS[146]. According to a recent meta-analysis, the combination of bevacizumab and FOLFOXIRI offers more promising results – the R0 surgery conversion rate was 28.1%, and the median OS and PFS were 30.2 and 12.4 mo respectively[147]. Multiple randomized trials have shown the addition of cetuximab to chemotherapy in KRAS wild-type (WT) unresectable disease improved the R0 resection rate by 2-3 folds[144,148]. An increase in complete resection rate from 11 to 18%, however, did not translate into survival benefit in a meta-analysis[149]. Panitumumab, another anti-EGFR agent, has also been linked with greater likelihood of curative resection when added to FOLFOX (29% vs 17%) in KRAS-WT unresectable CRLM[150].

Adding floxuridine-HAI to best systemic chemotherapy achieved a 47% conversion rate to resectable disease at a median of 6 months; median OS and PFS were 38 and 13 mo[151]. Oxaliplatin-HAI, meanwhile, was associated with response rates ranging from 24% to 81% in multiple small-scale studies[152]. In the French multicentric OPTILIV trial, triplet-agent-HAI plus systemic cetuximab gave a 30% secondary resection rate in KRAS-WT disease[153]. Whether HAI strategies offer additional benefit to modern intensive systemic chemotherapy regimens have to be tested by future RCTs.

Of note, patients who required prolonged chemotherapy (> 12 cycles) to achieve resectability had higher perioperative morbidity and inferior oncological outcomes. Conservative strategies rather than radical operation may be more appropriate for this subgroup[154]. Nonetheless, in general, conversion therapy followed by curative resection should be attempted whenever possible, as survival in this secondary R0 resected group is similar to those who had curative upfront surgery; early recurrence is not uncommon though.

LIVER-DIRECTED THERAPIES IN UNRESECTABLE CRLM

Clinicians nowadays can choose from an ever-growing armamentarium of loco-regional therapies to attain hepatic disease control. Yet, there is a lack of high-quality evidence assessing the benefits of each modality and no large-scale trials compared the different treatment strategies. Most of these liver-directed therapies require specific expertise, and should be recommended only in institutions with extensive experience with the procedure; the choice of treatment modality often depends on the availability of expertise. Here we will present an update of the available evidence regarding popular treatment modalities.

Ablation

Radiofrequency ablation (RFA) produces heat by delivering high-frequency alternating electric current through an electrode. Its widespread application has been limited by the relatively high local recurrence rate (ranged from 5% to 60% across different studies) and associated technical barriers e.g., tissue desiccation (charring) and heat-sink effect affect energy delivery[109]. Five-year survival varies between 17% and 51%[155]. According to an international expert panel position paper, thermal ablation is indicated for limited number (five or fewer) of small-sized (preferably < 3 cm) metastases deemed inoperable because of tumour or patient factors (e.g., multiple comorbidities); well-located tumours up to 5 cm or patients with up to nine tumours could also be considered[155]. Complete ablation with 10 mm margins in all directions should be attained[156]. In the landmark EORTC CLOCC study (40004), on top of systemic chemotherapy, RFA offered PFS benefit (median 16.8 mo vs 9.9 mo) and a significant prolongation of OS (median 45.6 mo vs 40.5 mo) in unresectable CRLM[157,158].

Producing heat from oscillation of water molecules, microwave ablation (MWA) is less susceptible to charring and heat sink, and could be more efficient in the treatment of large lesions and those near major hepatic vessels. It has been associated with lower local recurrence rate compared to RFA, with similar long-term survival outcomes and safety profiles[159,160].

Irreversible electroporation (IRE) induces cell death by creating permanent nanopores in cellular membranes while preserving tissue architecture. It can be a good ablative modality for tumours adjacent to major vascular or biliary structures. Early series showed encouraging results with PFS and OS rates of 18 and 62% at 2 years, but further evidence is necessary before advocation for its more widespread use in clinical practice[109,161].

Intra-arterial therapies

Intra-arterial therapies (IATs) are used to palliate symptoms or prolong survival in selected patients with unresectable CRLM refractory to chemotherapy. Selective infusion of tumoricidal and/or embolizing agents into hepatic artery branches enhances their delivery to liver tumours, while minimizes their effect on normal liver parenchyma; thereby avoiding significant hepatic and systemic toxicity.

Trans-arterial chemoembolization (TACE) kills cancer cells by means of high concentration of cytotoxic agents and ischemia. For CRLM, it is mainly used as rescue therapy for chemo-refractory diseases, although evidence for that is lacking. Mitomycin C- and cisplatin/doxorubicin-based conventional TACE (cTACE) offered median survivals of 14 and 11 months from the start of salvage therapy[162,163]. Post-embolization syndrome was reported in two-thirds of patients, but most only have nausea, fever, fatigue and mildly deranged liver function; severe complications e.g., liver abscess, hepatic failure and peptic ulcer were rare[164].

The newer drug-eluting bead (DEB)-TACE utilizes microspheres loaded with cytotoxics (doxorubicin or irinotecan); the drug is released in a controlled manner and a higher intra-tumoral dose can be delivered, meanwhile reduced peak plasma concentration may improve patient tolerance. In patients who failed previous chemotherapy, the median OS and PFS after DEB-TACE were 25 and 8 mo respectively[165]. In a small RCT, irinotecan-loaded DEB-TACE alone achieved better OS (median 22 mo vs 15 mo), PFS (7 mo vs 4 mo) and quality of life than systemic FOLFIRI; this needs to be verified in larger studies though[166]. Compared with cTACE, fewer patients (30%) experienced drug-related adverse events and most reported only minor symptoms e.g., abdominal pain, vomiting and fever[164]. Recent studies explored the combination of DEB-TACE with systemic chemotherapy or other treatment modalities. Concurrent capecitabine administration improved disease control but not survival[167], while adding FOLFOX and bevacizumab increased the conversion rate to resectability[168]. In non-surgical candidates, RFA combined with TACE attained local tumour control in 92% and OS at 2 years was 88.0%[169]. With better understanding of the properties of drug-eluting microspheres, results of DEB-TACE will improve and it will become a therapeutic option earlier in the course of disease e.g., neoadjuvant therapy, first-line treatment for unresectable disease.

For selective internal radiation therapy (SIRT), instead of cytotoxics, radiolabeled microspheres (Yttrium-90) were infused into the arterial system, delivering an effective dose of radiation to the tumour without causing intolerable toxicity to normal liver. Similar to TACE, it is typically considered in CRLM not amenable to resection or ablation. Earlier data confirmed its role in chemo-refractory disease – when given together with systemic 5-FU as salvage therapy, SIRT prolonged PFS (from 2.1 to 4.5 mo) in spite of no OS advantage[170]. However, more recent evidence did not support SIRT as a first-line therapy. Pooled data from 3 randomized trials (FOXFIRE, SIRFLOX and FOXFIRE-Global; including 1103 patients not suitable for curative resection or ablation and have not received any chemotherapy) showed, although associated with a higher objective response rate (72% vs 63%), early use of SIRT in combination with FOLFOX did not improve OS, PFS or hepatic resection rate, compared with FOLFOX alone (median OS 22.6 mo vs 23.3 mo, median PFS 11.1 vs 11.9 mo, hepatic resection rate 17% vs 16%). The SIRT group had more grade 3 or above adverse events (OR 1.42, 74% vs 67%). The authors concluded SIRT should not be used in unselected chemotherapy-naive liver-dominant or liver-only CRLM[171]. Further studies can help define patient selection and the role of SIRT in the management of CRLM.

In unresectable CRLM, HAI achieved a better tumour response rate but similar survival compared with standard 5-FU chemotherapy[172]; its role in the era of modern chemotherapy and targeted agents is less well-defined. HAI has been associated with relative high rates of technical failure (hepatic artery dissection and thrombosis 21%, catheter occlusion 5%, pump failure and infection 2% and 3%); together with the special equipment and expertise required, its availability is limited to relatively few centres[164].

No randomized trial has compared the efficacy and safety of the above three IAT modalities; evidence supporting one over the other is lacking. A recent systematic review tried to settle this issue - the pooled RECIST response rates for cTACE, DEB-TACE and SIRT were 23%, 36% and 23%, while medians survivals from first therapy were 16, 16 and 12 mo respectively[173]. However, significant heterogeneity in terms of patient characteristics, tumour burden, previous and post-IAT therapies exists between the included studies; and this precluded meaningful comparisons between the three therapies. We need higher-quality evidence to properly answer this question.

Stereotactic body radiation therapy

This technique allows delivery of a conformal high dose radiation to the tumour, while sparing normal liver. It is suitable for patients with adequate hepatic function but unresectable CRLM. Precise selection criteria are not well defined; some included good performance status, not more than 3 CRLMs and the largest tumour size less than 3 cm[174]. From a systematic review including 18 heterogenous studies with different RT doses and schedules (most patients had 1-2 oligo-metastases), the pooled 1- and 2-year local control rates were 67% and 59.3%, while one- and two-year OS were 67.2% and 56.5% respectively; mild/moderate and severe liver toxicity occurred in 30.7% and 8.7% patients[175]. The limited evidence so far showed encouraging results; however, this has to be validated in large prospective trials. Guidelines define stereotactic body radiation therapy as a reasonable treatment option for patients unsuitable for surgery or ablative therapies[9,10].

Controversy 6: Role of liver transplantation: Traditionally CRLM was regarded as a contraindication to liver transplantation (LT); this concept was challenged by the pilot SECA study[176]. Twenty-one patients with unresectable liver-only CRC metastases underwent deceased donor liver transplantation after at least 6 weeks of chemotherapy. OS rates of 95%, 68% and 60% at 1, 3 and 5 years were comparable to results of LT for other indications, and significantly better than a similar cohort who received first-line chemotherapy (5-year OS 9%). However, only 35% patients remained recurrence-free at 1 year; most of the recurrences were small slow-growing lung metastases. Compared with the chemotherapy group, the equivalent DFS but markedly superior OS attained by LT can be attributed to the different metastatic patterns – progression of non-resected LM carries a much worse prognosis than the post-LT indolent pulmonary metastases[177]. Similar OS rates were observed in a small series containing 12 patients – one third remained relapse-free after 4 years, suggesting LT may achieve long-term cure in selected patients with unresectable CRLM[178].

These results have to be interpreted with caution, though. They were small studies and there were no standardized selection criteria for recruiting these patients; it is questionable whether this survival benefit can be reproduced in other patients. Currently, LT remains experimental until better selection criteria help achieve lower recurrence rate, particularly in the setting of limited liver graft availability. A number of trials are underway to address the potential of LT for unresectable CRLM, including clarification of survival advantage by RCTs, the role of living donor LT, and the safety and efficacy of total hepatectomy after transplantation of left lateral section graft[179-182].

CONCLUSION

Recent advances in CRLM management have significantly improved outcome on the one hand while complicating the formulation of treatment strategy on the other. Multi-disciplinary involvement from the outset helps define resectability and devise personalized treatment approach. Refined patient selection, with greater emphasis on tumour biology, ensures patients benefit most from the offered interventions.

Surgical resection remains the cornerstone of treatment for curative intent. Liver augmentation strategies and conversion therapy have expanded the definition of resectability and increased the number of patients getting cured. The role of neoadjuvant therapy in operable disease is still controversial, while the use of adjuvant chemotherapy has gained generalized acceptance. Liver-directed therapies are getting more popular, resulting in better local disease control; however, they are currently not recommended as first-line treatment in unresectable CRLM. Liver transplant remains experimental and needs further evidence to validate its use. In the absence of standardized evidence-based protocols, the optimal management of CRLM should be determined by a multi-disciplinary team.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report classification

Grade A (Excellent): A, A

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Farshadpour F, Kaya M, Tijera FH S- Editor: Ma YJ L- Editor: A E- Editor: Zhang YL

References
1.  Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23762]  [Cited by in F6Publishing: 25159]  [Article Influence: 1935.3]  [Reference Citation Analysis (3)]
2.  van der Pool AE, Damhuis RA, Ijzermans JN, de Wilt JH, Eggermont AM, Kranse R, Verhoef C. Trends in incidence, treatment and survival of patients with stage IV colorectal cancer: a population-based series. Colorectal Dis. 2012;14:56-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 160]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
3.  Kanas GP, Taylor A, Primrose JN, Langeberg WJ, Kelsh MA, Mowat FS, Alexander DD, Choti MA, Poston G. Survival after liver resection in metastatic colorectal cancer: review and meta-analysis of prognostic factors. Clin Epidemiol. 2012;4:283-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 258]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
4.  Tomlinson JS, Jarnagin WR, DeMatteo RP, Fong Y, Kornprat P, Gonen M, Kemeny N, Brennan MF, Blumgart LH, D'Angelica M. Actual 10-year survival after resection of colorectal liver metastases defines cure. J Clin Oncol. 2007;25:4575-4580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 845]  [Cited by in F6Publishing: 845]  [Article Influence: 49.7]  [Reference Citation Analysis (0)]
5.  Jones RP, Jackson R, Dunne DF, Malik HZ, Fenwick SW, Poston GJ, Ghaneh P. Systematic review and meta-analysis of follow-up after hepatectomy for colorectal liver metastases. Br J Surg. 2012;99:477-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 123]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
6.  Weledji EP. Centralization of Liver Cancer Surgery and Impact on Multidisciplinary Teams Working on Stage IV Colorectal Cancer. Oncol Rev. 2017;11:331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 16]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
7.  Adam R, de Gramont A, Figueras J, Kokudo N, Kunstlinger F, Loyer E, Poston G, Rougier P, Rubbia-Brandt L, Sobrero A, Teh C, Tejpar S, Van Cutsem E, Vauthey JN, Påhlman L; of the EGOSLIM (Expert Group on OncoSurgery management of LIver Metastases) group. Managing synchronous liver metastases from colorectal cancer: a multidisciplinary international consensus. Cancer Treat Rev. 2015;41:729-741.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 287]  [Cited by in F6Publishing: 336]  [Article Influence: 37.3]  [Reference Citation Analysis (0)]
8.  Oxenberg J, Papenfuss W, Esemuede I, Attwood K, Simunovic M, Kuvshinoff B, Francescutti V. Multidisciplinary cancer conferences for gastrointestinal malignancies result in measureable treatment changes: a prospective study of 149 consecutive patients. Ann Surg Oncol. 2015;22:1533-1539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
9.  Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, Aranda Aguilar E, Bardelli A, Benson A, Bodoky G, Ciardiello F, D'Hoore A, Diaz-Rubio E, Douillard JY, Ducreux M, Falcone A, Grothey A, Gruenberger T, Haustermans K, Heinemann V, Hoff P, Köhne CH, Labianca R, Laurent-Puig P, Ma B, Maughan T, Muro K, Normanno N, Österlund P, Oyen WJ, Papamichael D, Pentheroudakis G, Pfeiffer P, Price TJ, Punt C, Ricke J, Roth A, Salazar R, Scheithauer W, Schmoll HJ, Tabernero J, Taïeb J, Tejpar S, Wasan H, Yoshino T, Zaanan A, Arnold D. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27:1386-1422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2286]  [Cited by in F6Publishing: 2145]  [Article Influence: 268.1]  [Reference Citation Analysis (0)]
10.  National Comprehensive Cancer Network. NCCN clinical practice in oncology: colon cancer. NCCN.org 2018, version 3.  Available from: https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Rojas Llimpe FL, Di Fabio F, Ercolani G, Giampalma E, Cappelli A, Serra C, Castellucci P, D'Errico A, Golfieri R, Pinna AD, Pinto C. Imaging in resectable colorectal liver metastasis patients with or without preoperative chemotherapy: results of the PROMETEO-01 study. Br J Cancer. 2014;111:667-673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 47]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
12.  Kulemann V, Schima W, Tamandl D, Kaczirek K, Gruenberger T, Wrba F, Weber M, Ba-Ssalamah A. Preoperative detection of colorectal liver metastases in fatty liver: MDCT or MRI? Eur J Radiol. 2011;79:e1-e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 71]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
13.  Zech CJ, Korpraphong P, Huppertz A, Denecke T, Kim MJ, Tanomkiat W, Jonas E, Ba-Ssalamah A; VALUE study group. Randomized multicentre trial of gadoxetic acid-enhanced MRI versus conventional MRI or CT in the staging of colorectal cancer liver metastases. Br J Surg. 2014;101:613-621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 102]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
14.  Ruers TJ, Wiering B, van der Sijp JR, Roumen RM, de Jong KP, Comans EF, Pruim J, Dekker HM, Krabbe PF, Oyen WJ. Improved selection of patients for hepatic surgery of colorectal liver metastases with (18)F-FDG PET: a randomized study. J Nucl Med. 2009;50:1036-1041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 137]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
15.  Moulton CA, Gu CS, Law CH, Tandan VR, Hart R, Quan D, Fairfull Smith RJ, Jalink DW, Husien M, Serrano PE, Hendler AL, Haider MA, Ruo L, Gulenchyn KY, Finch T, Julian JA, Levine MN, Gallinger S. Effect of PET before liver resection on surgical management for colorectal adenocarcinoma metastases: a randomized clinical trial. JAMA. 2014;311:1863-1869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 125]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
16.  Serrano PES, Gu CS, Husien M, Jalink D, Martel G, Tsang ME, Hallet JI, Gallinger S, Ritter A, McAlister V, Sela N, Solomon H, Beyfuss K, Li C, Lee E, Moulton CA, Levine MN. Effect of PET-CT on disease recurrence and its management in patients with potentially resectable colorectal cancer liver metastases: The long-term results of a randomized controlled trial (PET-CT Imaging prior to liver resection for colorectal adenocarcinoma metastases). J Clin Oncol. 2018;36:15_suppl, 3527.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Maffione AM, Lopci E, Bluemel C, Giammarile F, Herrmann K, Rubello D. Diagnostic accuracy and impact on management of (18)F-FDG PET and PET/CT in colorectal liver metastasis: a meta-analysis and systematic review. Eur J Nucl Med Mol Imaging. 2015;42:152-163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 97]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
18.  Lake ES, Wadhwani S, Subar D, Kauser A, Harris C, Chang D, Lapsia S. The influence of FDG PET-CT on the detection of extrahepatic disease in patients being considered for resection of colorectal liver metastasis. Ann R Coll Surg Engl. 2014;96:211-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
19.  Cieslak KP, Runge JH, Heger M, Stoker J, Bennink RJ, van Gulik TM. New perspectives in the assessment of future remnant liver. Dig Surg. 2014;31:255-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 59]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
20.  Lau H, Man K, Fan ST, Yu WC, Lo CM, Wong J. Evaluation of preoperative hepatic function in patients with hepatocellular carcinoma undergoing hepatectomy. Br J Surg. 1997;84:1255-1259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
21.  De Gasperi A, Mazza E, Prosperi M. Indocyanine green kinetics to assess liver function: Ready for a clinical dynamic assessment in major liver surgery? World J Hepatol. 2016;8:355-367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 103]  [Article Influence: 12.9]  [Reference Citation Analysis (4)]
22.  de Graaf W, van Lienden KP, Dinant S, Roelofs JJ, Busch OR, Gouma DJ, Bennink RJ, van Gulik TM. Assessment of future remnant liver function using hepatobiliary scintigraphy in patients undergoing major liver resection. J Gastrointest Surg. 2010;14:369-378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 181]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
23.  Adams RB, Aloia TA, Loyer E, Pawlik TM, Taouli B, Vauthey JN; Americas Hepato-Pancreato-Biliary Association; Society of Surgical Oncology; Society for Surgery of the Alimentary Tract. Selection for hepatic resection of colorectal liver metastases: expert consensus statement. HPB (Oxford). 2013;15:91-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 211]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
24.  Ribero D, Viganò L, Amisano M, Capussotti L. Prognostic factors after resection of colorectal liver metastases: from morphology to biology. Future Oncol. 2013;9:45-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
25.  Jones RP, Brudvik KW, Franklin JM, Poston GJ. Precision surgery for colorectal liver metastases: Opportunities and challenges of omics-based decision making. Eur J Surg Oncol. 2017;43:875-883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 23]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
26.  Sasaki K, Andreatos N, Margonis GA, He J, Weiss M, Johnston F, Wolfgang C, Antoniou E, Pikoulis E, Pawlik TM. The prognostic implications of primary colorectal tumor location on recurrence and overall survival in patients undergoing resection for colorectal liver metastasis. J Surg Oncol. 2016;114:803-809.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 61]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
27.  Fong Y, Fortner J, Sun RL, Brennan MF, Blumgart LH. Clinical score for predicting recurrence after hepatic resection for metastatic colorectal cancer: analysis of 1001 consecutive cases. Ann Surg. 1999;230:309-18; discussion 318-21.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Roberts KJ, White A, Cockbain A, Hodson J, Hidalgo E, Toogood GJ, Lodge JP. Performance of prognostic scores in predicting long-term outcome following resection of colorectal liver metastases. Br J Surg. 2014;101:856-866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 60]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
29.  Kumar R, Dennison AR, Robertson V, Jones MJ, Neal CP, Garcea G. Clinical risk scores in the current era of neoadjuvant chemotherapy for colorectal liver metastases. ANZ J Surg. 2018;88:E16-E20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
30.  Blazer DG, Kishi Y, Maru DM, Kopetz S, Chun YS, Overman MJ, Fogelman D, Eng C, Chang DZ, Wang H, Zorzi D, Ribero D, Ellis LM, Glover KY, Wolff RA, Curley SA, Abdalla EK, Vauthey JN. Pathologic response to preoperative chemotherapy: a new outcome end point after resection of hepatic colorectal metastases. J Clin Oncol. 2008;26:5344-5351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 448]  [Cited by in F6Publishing: 475]  [Article Influence: 29.7]  [Reference Citation Analysis (0)]
31.  Shindoh J, Loyer EM, Kopetz S, Boonsirikamchai P, Maru DM, Chun YS, Zimmitti G, Curley SA, Charnsangavej C, Aloia TA, Vauthey JN. Optimal morphologic response to preoperative chemotherapy: an alternate outcome end point before resection of hepatic colorectal metastases. J Clin Oncol. 2012;30:4566-4572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 154]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
32.  Giessen C, Laubender RP, Fischer von Weikersthal L, Schalhorn A, Modest DP, Stintzing S, Haas M, Mansmann UR, Heinemann V. Early tumor shrinkage in metastatic colorectal cancer: retrospective analysis from an irinotecan-based randomized first-line trial. Cancer Sci. 2013;104:718-724.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
33.  Modest DP, Laubender RP, Stintzing S, Giessen C, Schulz C, Haas M, Mansmann U, Heinemann V. Early tumor shrinkage in patients with metastatic colorectal cancer receiving first-line treatment with cetuximab combined with either CAPIRI or CAPOX: an analysis of the German AIO KRK 0104 trial. Acta Oncol. 2013;52:956-962.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 49]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
34.  Xia Q, Liu J, Wu C, Song S, Tong L, Huang G, Feng Y, Jiang Y, Liu Y, Yin T, Ni Y. Prognostic significance of (18)FDG PET/CT in colorectal cancer patients with liver metastases: a meta-analysis. Cancer Imaging. 2015;15:19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
35.  Adam R, Wicherts DA, de Haas RJ, Aloia T, Lévi F, Paule B, Guettier C, Kunstlinger F, Delvart V, Azoulay D, Castaing D. Complete pathologic response after preoperative chemotherapy for colorectal liver metastases: myth or reality? J Clin Oncol. 2008;26:1635-1641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 196]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
36.  Knijn N, Mekenkamp LJ, Klomp M, Vink-Börger ME, Tol J, Teerenstra S, Meijer JW, Tebar M, Riemersma S, van Krieken JH, Punt CJ, Nagtegaal ID. KRAS mutation analysis: a comparison between primary tumours and matched liver metastases in 305 colorectal cancer patients. Br J Cancer. 2011;104:1020-1026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 227]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
37.  Zimmitti G, Shindoh J, Mise Y, Kopetz S, Loyer EM, Andreou A, Cooper AB, Kaur H, Aloia TA, Maru DM, Vauthey JN. RAS mutations predict radiologic and pathologic response in patients treated with chemotherapy before resection of colorectal liver metastases. Ann Surg Oncol. 2015;22:834-842.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 75]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
38.  Brudvik KW, Kopetz SE, Li L, Conrad C, Aloia TA, Vauthey JN. Meta-analysis of KRAS mutations and survival after resection of colorectal liver metastases. Br J Surg. 2015;102:1175-1183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 138]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
39.  Passot G, Denbo JW, Yamashita S, Kopetz SE, Chun YS, Maru D, Overman MJ, Brudvik KW, Conrad C, Aloia TA, Vauthey JN. Is hepatectomy justified for patients with RAS mutant colorectal liver metastases? An analysis of 524 patients undergoing curative liver resection. Surgery. 2017;161:332-340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 31]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
40.  Brudvik KW, Jones RP, Giuliante F, Shindoh J, Passot G, Chung MH, Song J, Li L, Dagenborg VJ, Fretland ÅA, Røsok B, De Rose AM, Ardito F, Edwin B, Panettieri E, Larocca LM, Yamashita S, Conrad C, Aloia TA, Poston GJ, Bjørnbeth BA, Vauthey JN. RAS Mutation Clinical Risk Score to Predict Survival After Resection of Colorectal Liver Metastases. Ann Surg. 2017;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 130]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
41.  Løes IM, Immervoll H, Sorbye H, Angelsen JH, Horn A, Knappskog S, Lønning PE. Impact of KRAS, BRAF, PIK3CA, TP53 status and intraindividual mutation heterogeneity on outcome after liver resection for colorectal cancer metastases. Int J Cancer. 2016;139:647-656.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 61]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
42.  Ursem C, Atreya CE, Van Loon K. Emerging treatment options for BRAF-mutant colorectal cancer. Gastrointest Cancer. 2018;8:13-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 31]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
43.  Balachandran VP, Arora A, Gönen M, Ito H, Turcotte S, Shia J, Viale A, Snoeren N, van Hooff SR, Rinkes IH, Adam R, Kingham TP, Allen PJ, DeMatteo RP, Jarnagin WR, D'Angelica MI. A Validated Prognostic Multigene Expression Assay for Overall Survival in Resected Colorectal Cancer Liver Metastases. Clin Cancer Res. 2016;22:2575-2582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 29]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
44.  Seeberg LT, Waage A, Brunborg C, Hugenschmidt H, Renolen A, Stav I, Bjørnbeth BA, Brudvik KW, Borgen EF, Naume B, Wiedswang G. Circulating tumor cells in patients with colorectal liver metastasis predict impaired survival. Ann Surg. 2015;261:164-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 47]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
45.  Mostert B, Jiang Y, Sieuwerts AM, Wang H, Bolt-de Vries J, Biermann K, Kraan J, Lalmahomed Z, van Galen A, de Weerd V, van der Spoel P, Ramírez-Moreno R, Verhoef C, Ijzermans JN, Wang Y, Gratama JW, Foekens JA, Sleijfer S, Martens JW. KRAS and BRAF mutation status in circulating colorectal tumor cells and their correlation with primary and metastatic tumor tissue. Int J Cancer. 2013;133:130-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 115]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
46.  El Messaoudi S, Mouliere F, Du Manoir S, Bascoul-Mollevi C, Gillet B, Nouaille M, Fiess C, Crapez E, Bibeau F, Theillet C, Mazard T, Pezet D, Mathonnet M, Ychou M, Thierry AR. Circulating DNA as a Strong Multimarker Prognostic Tool for Metastatic Colorectal Cancer Patient Management Care. Clin Cancer Res. 2016;22:3067-3077.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 114]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
47.  Zarour LR, Anand S, Billingsley KG, Bisson WH, Cercek A, Clarke MF, Coussens LM, Gast CE, Geltzeiler CB, Hansen L, Kelley KA, Lopez CD, Rana SR, Ruhl R, Tsikitis VL, Vaccaro GM, Wong MH, Mayo SC. Colorectal Cancer Liver Metastasis: Evolving Paradigms and Future Directions. Cell Mol Gastroenterol Hepatol. 2017;3:163-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 181]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
48.  Bedin C, Enzo MV, Del Bianco P, Pucciarelli S, Nitti D, Agostini M. Diagnostic and prognostic role of cell-free DNA testing for colorectal cancer patients. Int J Cancer. 2017;140:1888-1898.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 73]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
49.  Hur K, Toiyama Y, Schetter AJ, Okugawa Y, Harris CC, Boland CR, Goel A. Identification of a metastasis-specific MicroRNA signature in human colorectal cancer. J Natl Cancer Inst. 2015;107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 124]  [Article Influence: 13.8]  [Reference Citation Analysis (1)]
50.  Chua TC, Saxena A, Liauw W, Kokandi A, Morris DL. Systematic review of randomized and nonrandomized trials of the clinical response and outcomes of neoadjuvant systemic chemotherapy for resectable colorectal liver metastases. Ann Surg Oncol. 2010;17:492-501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 112]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
51.  Nigri G, Petrucciani N, Ferla F, La Torre M, Aurello P, Ramacciato G. Neoadjuvant chemotherapy for resectable colorectal liver metastases: what is the evidence? Results of a systematic review of comparative studies. Surgeon. 2015;13:83-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
52.  Nordlinger B, Sorbye H, Glimelius B, Poston GJ, Schlag PM, Rougier P, Bechstein WO, Primrose JN, Walpole ET, Finch-Jones M, Jaeck D, Mirza D, Parks RW, Collette L, Praet M, Bethe U, Van Cutsem E, Scheithauer W, Gruenberger T; EORTC Gastro-Intestinal Tract Cancer Group; Cancer Research UK; Arbeitsgruppe Lebermetastasen und-tumoren in der Chirurgischen Arbeitsgemeinschaft Onkologie (ALM-CAO); Australasian Gastro-Intestinal Trials Group (AGITG); Fédération Francophone de Cancérologie Digestive (FFCD). Perioperative chemotherapy with FOLFOX4 and surgery versus surgery alone for resectable liver metastases from colorectal cancer (EORTC Intergroup trial 40983): a randomised controlled trial. Lancet. 2008;371:1007-1016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1478]  [Cited by in F6Publishing: 1380]  [Article Influence: 86.3]  [Reference Citation Analysis (0)]
53.  Nordlinger B, Sorbye H, Glimelius B, Poston GJ, Schlag PM, Rougier P, Bechstein WO, Primrose JN, Walpole ET, Finch-Jones M, Jaeck D, Mirza D, Parks RW, Mauer M, Tanis E, Van Cutsem E, Scheithauer W, Gruenberger T; EORTC Gastro-Intestinal Tract Cancer Group; Cancer Research UK; Arbeitsgruppe Lebermetastasen und–tumoren in der Chirurgischen Arbeitsgemeinschaft Onkologie (ALM-CAO); Australasian Gastro-Intestinal Trials Group (AGITG); Fédération Francophone de Cancérologie Digestive (FFCD). Perioperative FOLFOX4 chemotherapy and surgery versus surgery alone for resectable liver metastases from colorectal cancer (EORTC 40983): long-term results of a randomised, controlled, phase 3 trial. Lancet Oncol. 2013;14:1208-1215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 760]  [Cited by in F6Publishing: 823]  [Article Influence: 74.8]  [Reference Citation Analysis (0)]
54.  Liu W, Zhou JG, Sun Y, Zhang L, Xing BC. The role of neoadjuvant chemotherapy for resectable colorectal liver metastases: a systematic review and meta-analysis. Oncotarget. 2016;7:37277-37287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
55.  Ayez N, van der Stok EP, de Wilt H, Radema SA, van Hillegersberg R, Roumen RM, Vreugdenhil G, Tanis PJ, Punt CJ, Dejong CH, Jansen RL, Verheul HM, de Jong KP, Hospers GA, Klaase JM, Legdeur MC, van Meerten E, Eskens FA, van der Meer N, van der Holt B, Verhoef C, Grünhagen DJ. Neo-adjuvant chemotherapy followed by surgery versus surgery alone in high-risk patients with resectable colorectal liver metastases: the CHARISMA randomized multicenter clinical trial. BMC Cancer. 2015;15:180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 44]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
56.  Sabanathan D, Eslick GD, Shannon J. Use of Neoadjuvant Chemotherapy Plus Molecular Targeted Therapy in Colorectal Liver Metastases: A Systematic Review and Meta-analysis. Clin Colorectal Cancer. 2016;15:e141-e147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 17]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
57.  Primrose J, Falk S, Finch-Jones M, Valle J, O'Reilly D, Siriwardena A, Hornbuckle J, Peterson M, Rees M, Iveson T, Hickish T, Butler R, Stanton L, Dixon E, Little L, Bowers M, Pugh S, Garden OJ, Cunningham D, Maughan T, Bridgewater J. Systemic chemotherapy with or without cetuximab in patients with resectable colorectal liver metastasis: the New EPOC randomised controlled trial. Lancet Oncol. 2014;15:601-611.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 325]  [Cited by in F6Publishing: 293]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
58.  Nasti G, Piccirillo MC, Izzo F, Ottaiano A, Albino V, Delrio P, Romano C, Giordano P, Lastoria S, Caracò C, de Lutio di Castelguidone E, Palaia R, Daniele G, Aloj L, Romano G, Iaffaioli RV. Neoadjuvant FOLFIRI+bevacizumab in patients with resectable liver metastases from colorectal cancer: a phase 2 trial. Br J Cancer. 2013;108:1566-1570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 30]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
59.  Stein A, Glockzin G, Wienke A, Arnold D, Edelmann T, Hildebrandt B, Hollerbach S, Illerhaus G, Königsrainer A, Richter M, Schlitt HJ, Schmoll HJ. Treatment with bevacizumab and FOLFOXIRI in patients with advanced colorectal cancer: presentation of two novel trials (CHARTA and PERIMAX) and review of the literature. BMC Cancer. 2012;12:356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
60.  Viganò L, Capussotti L, De Rosa G, De Saussure WO, Mentha G, Rubbia-Brandt L. Liver resection for colorectal metastases after chemotherapy: impact of chemotherapy-related liver injuries, pathological tumor response, and micrometastases on long-term survival. Ann Surg. 2013;258:731-40; discussion 741-2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 138]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
61.  Zhao J, van Mierlo KMC, Gómez-Ramírez J, Kim H, Pilgrim CHC, Pessaux P, Rensen SS, van der Stok EP, Schaap FG, Soubrane O, Takamoto T, Viganò L, Winkens B, Dejong CHC, Olde Damink SWM; Chemotherapy-Associated Liver Injury (CALI) consortium. Systematic review of the influence of chemotherapy-associated liver injury on outcome after partial hepatectomy for colorectal liver metastases. Br J Surg. 2017;104:990-1002.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 61]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
62.  Welsh FK, Tilney HS, Tekkis PP, John TG, Rees M. Safe liver resection following chemotherapy for colorectal metastases is a matter of timing. Br J Cancer. 2007;96:1037-1042.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 159]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
63.  Kishi Y, Zorzi D, Contreras CM, Maru DM, Kopetz S, Ribero D, Motta M, Ravarino N, Risio M, Curley SA, Abdalla EK, Capussotti L, Vauthey JN. Extended preoperative chemotherapy does not improve pathologic response and increases postoperative liver insufficiency after hepatic resection for colorectal liver metastases. Ann Surg Oncol. 2010;17:2870-2876.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 168]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
64.  Robinson PJ. The effects of cancer chemotherapy on liver imaging. Eur Radiol. 2009;19:1752-1762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 64]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
65.  van Kessel CS, Buckens CF, van den Bosch MA, van Leeuwen MS, van Hillegersberg R, Verkooijen HM. Preoperative imaging of colorectal liver metastases after neoadjuvant chemotherapy: a meta-analysis. Ann Surg Oncol. 2012;19:2805-2813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 127]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
66.  Kuhlmann K, van Hilst J, Fisher S, Poston G. Management of disappearing colorectal liver metastases. Eur J Surg Oncol. 2016;42:1798-1805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 19]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
67.  Benoist S, Brouquet A, Penna C, Julié C, El Hajjam M, Chagnon S, Mitry E, Rougier P, Nordlinger B. Complete response of colorectal liver metastases after chemotherapy: does it mean cure? J Clin Oncol. 2006;24:3939-3945.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 423]  [Cited by in F6Publishing: 370]  [Article Influence: 20.6]  [Reference Citation Analysis (0)]
68.  Zendel A, Lahat E, Dreznik Y, Zakai BB, Eshkenazy R, Ariche A. "Vanishing liver metastases"-A real challenge for liver surgeons. Hepatobiliary Surg Nutr. 2014;3:295-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 10]  [Reference Citation Analysis (0)]
69.  van Vledder MG, de Jong MC, Pawlik TM, Schulick RD, Diaz LA, Choti MA. Disappearing colorectal liver metastases after chemotherapy: should we be concerned? J Gastrointest Surg. 2010;14:1691-1700.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 89]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
70.  Bischof DA, Clary BM, Maithel SK, Pawlik TM. Surgical management of disappearing colorectal liver metastases. Br J Surg. 2013;100:1414-1420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 52]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
71.  Ntourakis D, Memeo R, Soler L, Marescaux J, Mutter D, Pessaux P. Augmented Reality Guidance for the Resection of Missing Colorectal Liver Metastases: An Initial Experience. World J Surg. 2016;40:419-426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 55]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
72.  van der Geest LG, Lam-Boer J, Koopman M, Verhoef C, Elferink MA, de Wilt JH. Nationwide trends in incidence, treatment and survival of colorectal cancer patients with synchronous metastases. Clin Exp Metastasis. 2015;32:457-465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 264]  [Cited by in F6Publishing: 324]  [Article Influence: 36.0]  [Reference Citation Analysis (0)]
73.  Bonney GK, Coldham C, Adam R, Kaiser G, Barroso E, Capussotti L, Laurent C, Verhoef C, Nuzzo G, Elias D, Lapointe R, Hubert C, Lopez-Ben S, Krawczyk M, Mirza DF; LiverMetSurvey International Registry Working Group. Role of neoadjuvant chemotherapy in resectable synchronous colorectal liver metastasis; An international multi-center data analysis using LiverMetSurvey. J Surg Oncol. 2015;111:716-724.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 39]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
74.  Wanis KN, Pineda-Solis K, Tun-Abraham ME, Yeoman J, Welch S, Vogt K, Van Koughnett JAM, Ott M, Hernandez-Alejandro R. Management of colorectal cancer with synchronous liver metastases: impact of multidisciplinary case conference review. Hepatobiliary Surg Nutr. 2017;6:162-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
75.  Andres A, Toso C, Adam R, Barroso E, Hubert C, Capussotti L, Gerstel E, Roth A, Majno PE, Mentha G. A survival analysis of the liver-first reversed management of advanced simultaneous colorectal liver metastases: a LiverMetSurvey-based study. Ann Surg. 2012;256:772-8; discussion 778-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 81]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
76.  Waisberg J, Ivankovics IG. Liver-first approach of colorectal cancer with synchronous hepatic metastases: A reverse strategy. World J Hepatol. 2015;7:1444-1449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 15]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
77.  Lam VW, Laurence JM, Pang T, Johnston E, Hollands MJ, Pleass HC, Richardson AJ. A systematic review of a liver-first approach in patients with colorectal cancer and synchronous colorectal liver metastases. HPB (Oxford). 2014;16:101-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 50]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
78.  de Jong MC, Beckers RCJ, van Woerden V, Sijmons JML, Bemelmans MHA, van Dam RM, Dejong CHC. The liver-first approach for synchronous colorectal liver metastases: more than a decade of experience in a single centre. HPB (Oxford). 2018;20:631-640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
79.  Silberhumer GR, Paty PB, Temple LK, Araujo RL, Denton B, Gonen M, Nash GM, Allen PJ, DeMatteo RP, Guillem J, Weiser MR, D'Angelica MI, Jarnagin WR, Wong DW, Fong Y. Simultaneous resection for rectal cancer with synchronous liver metastasis is a safe procedure. Am J Surg. 2015;209:935-942.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 73]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
80.  Feo L, Polcino M, Nash GM. Resection of the Primary Tumor in Stage IV Colorectal Cancer: When Is It Necessary? Surg Clin North Am. 2017;97:657-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
81.  Mayo SC, Pulitano C, Marques H, Lamelas J, Wolfgang CL, de Saussure W, Choti MA, Gindrat I, Aldrighetti L, Barrosso E, Mentha G, Pawlik TM. Surgical management of patients with synchronous colorectal liver metastasis: a multicenter international analysis. J Am Coll Surg. 2013;216:707-16; discussion 716-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 116]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
82.  Silberhumer GR, Paty PB, Denton B, Guillem J, Gonen M, Araujo RLC, Nash GM, Temple LK, Allen PJ, DeMatteo RP, Weiser MR, Wong WD, Jarnagin WR, D'Angelica MI, Fong Y. Long-term oncologic outcomes for simultaneous resection of synchronous metastatic liver and primary colorectal cancer. Surgery. 2016;160:67-73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 59]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
83.  Gavriilidis P, Sutcliffe RP, Hodson J, Marudanayagam R, Isaac J, Azoulay D, Roberts KJ. Simultaneous versus delayed hepatectomy for synchronous colorectal liver metastases: a systematic review and meta-analysis. HPB (Oxford). 2018;20:11-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 40]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
84.  Kelly ME, Spolverato G, Lê GN, Mavros MN, Doyle F, Pawlik TM, Winter DC. Synchronous colorectal liver metastasis: a network meta-analysis review comparing classical, combined, and liver-first surgical strategies. J Surg Oncol. 2015;111:341-351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 104]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
85.  Clancy C, Burke JP, Barry M, Kalady MF, Calvin Coffey J. A meta-analysis to determine the effect of primary tumor resection for stage IV colorectal cancer with unresectable metastases on patient survival. Ann Surg Oncol. 2014;21:3900-3908.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 96]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
86.  Wurster EF, Tenckhoff S, Probst P, Jensen K, Dölger E, Knebel P, Diener MK, Büchler MW, Ulrich A. A systematic review and meta-analysis of the utility of repeated versus single hepatic resection for colorectal cancer liver metastases. HPB (Oxford). 2017;19:491-497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
87.  Moris D, Ronnekleiv-Kelly S, Rahnemai-Azar AA, Felekouras E, Dillhoff M, Schmidt C, Pawlik TM. Parenchymal-Sparing Versus Anatomic Liver Resection for Colorectal Liver Metastases: a Systematic Review. J Gastrointest Surg. 2017;21:1076-1085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 81]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
88.  Mise Y, Aloia TA, Brudvik KW, Schwarz L, Vauthey JN, Conrad C. Parenchymal-sparing Hepatectomy in Colorectal Liver Metastasis Improves Salvageability and Survival. Ann Surg. 2016;263:146-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 178]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
89.  Fretland ÅA, Dagenborg VJ, Bjørnelv GMW, Kazaryan AM, Kristiansen R, Fagerland MW, Hausken J, Tønnessen TI, Abildgaard A, Barkhatov L, Yaqub S, Røsok BI, Bjørnbeth BA, Andersen MH, Flatmark K, Aas E, Edwin B. Laparoscopic Versus Open Resection for Colorectal Liver Metastases: The OSLO-COMET Randomized Controlled Trial. Ann Surg. 2018;267:199-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 335]  [Cited by in F6Publishing: 408]  [Article Influence: 81.6]  [Reference Citation Analysis (0)]
90.  Zhang XL, Liu RF, Zhang D, Zhang YS, Wang T. Laparoscopic versus open liver resection for colorectal liver metastases: A systematic review and meta-analysis of studies with propensity score-based analysis. Int J Surg. 2017;44:191-203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 34]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
91.  Cherqui D, Wakabayashi G, Geller DA, Buell JF, Han HS, Soubrane O, O'Rourke N; International Laparoscopic Liver Society. The need for organization of laparoscopic liver resection. J Hepatobiliary Pancreat Sci. 2016;23:665-667.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
92.  Pawlik TM, Scoggins CR, Zorzi D, Abdalla EK, Andres A, Eng C, Curley SA, Loyer EM, Muratore A, Mentha G, Capussotti L, Vauthey JN. Effect of surgical margin status on survival and site of recurrence after hepatic resection for colorectal metastases. Ann Surg. 2005;241:715-722, discussion 722-discussion 724.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Sadot E, Groot Koerkamp B, Leal JN, Shia J, Gonen M, Allen PJ, DeMatteo RP, Kingham TP, Kemeny N, Blumgart LH, Jarnagin WR, DʼAngelica MI. Resection margin and survival in 2368 patients undergoing hepatic resection for metastatic colorectal cancer: surgical technique or biologic surrogate? Ann Surg. 2015;262:476-85; discussion 483-5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 129]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
94.  Hamady ZZ, Lodge JP, Welsh FK, Toogood GJ, White A, John T, Rees M. One-millimeter cancer-free margin is curative for colorectal liver metastases: a propensity score case-match approach. Ann Surg. 2014;259:543-548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 110]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
95.  Andreou A, Aloia TA, Brouquet A, Dickson PV, Zimmitti G, Maru DM, Kopetz S, Loyer EM, Curley SA, Abdalla EK, Vauthey JN. Margin status remains an important determinant of survival after surgical resection of colorectal liver metastases in the era of modern chemotherapy. Ann Surg. 2013;257:1079-1088.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 144]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
96.  Margonis GA, Spolverato G, Kim Y, Ejaz A, Pawlik TM. Intraoperative surgical margin re-resection for colorectal liver metastasis: is it worth the effort? J Gastrointest Surg. 2015;19:699-707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
97.  Qadan M, D'Angelica MI. Extending the Limits of Resection for Colorectal Liver Metastases: Positive Resection Margin and Outcome After Resection of Colorectal Cancer Liver Metastases. J Gastrointest Surg. 2017;21:196-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
98.  Watanabe T, Muro K, Ajioka Y, Hashiguchi Y, Ito Y, Saito Y, Hamaguchi T, Ishida H, Ishiguro M, Ishihara S, Kanemitsu Y, Kawano H, Kinugasa Y, Kokudo N, Murofushi K, Nakajima T, Oka S, Sakai Y, Tsuji A, Uehara K, Ueno H, Yamazaki K, Yoshida M, Yoshino T, Boku N, Fujimori T, Itabashi M, Koinuma N, Morita T, Nishimura G, Sakata Y, Shimada Y, Takahashi K, Tanaka S, Tsuruta O, Yamaguchi T, Yamaguchi N, Tanaka T, Kotake K, Sugihara K; Japanese Society for Cancer of the Colon and Rectum. Japanese Society for Cancer of the Colon and Rectum (JSCCR) guidelines 2016 for the treatment of colorectal cancer. Int J Clin Oncol. 2018;23:1-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 576]  [Cited by in F6Publishing: 564]  [Article Influence: 94.0]  [Reference Citation Analysis (0)]
99.  Hwang M, Jayakrishnan TT, Green DE, George B, Thomas JP, Groeschl RT, Erickson B, Pappas SG, Gamblin TC, Turaga KK. Systematic review of outcomes of patients undergoing resection for colorectal liver metastases in the setting of extra hepatic disease. Eur J Cancer. 2014;50:1747-1757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 53]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
100.  Hadden WJ, de Reuver PR, Brown K, Mittal A, Samra JS, Hugh TJ. Resection of colorectal liver metastases and extra-hepatic disease: a systematic review and proportional meta-analysis of survival outcomes. HPB (Oxford). 2016;18:209-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
101.  Andres A, Mentha G, Adam R, Gerstel E, Skipenko OG, Barroso E, Lopez-Ben S, Hubert C, Majno PE, Toso C. Surgical management of patients with colorectal cancer and simultaneous liver and lung metastases. Br J Surg. 2015;102:691-699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 76]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
102.  Mise Y, Kopetz S, Mehran RJ, Aloia TA, Conrad C, Brudvik KW, Taggart MW, Vauthey JN. Is complete liver resection without resection of synchronous lung metastases justified? Ann Surg Oncol. 2015;22:1585-1592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 51]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
103.  Leung U, Gönen M, Allen PJ, Kingham TP, DeMatteo RP, Jarnagin WR, D'Angelica MI. Colorectal Cancer Liver Metastases and Concurrent Extrahepatic Disease Treated With Resection. Ann Surg. 2017;265:158-165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 97]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
104.  Maggiori L, Goéré D, Viana B, Tzanis D, Dumont F, Honoré C, Eveno C, Elias D. Should patients with peritoneal carcinomatosis of colorectal origin with synchronous liver metastases be treated with a curative intent? A case-control study. Ann Surg. 2013;258:116-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 66]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
105.  Adam R, de Haas RJ, Wicherts DA, Vibert E, Salloum C, Azoulay D, Castaing D. Concomitant extrahepatic disease in patients with colorectal liver metastases: when is there a place for surgery? Ann Surg. 2011;253:349-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 115]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
106.  Loveman E, Jones J, Clegg AJ, Picot J, Colquitt JL, Mendes D, Breen DJ, Moore E, George S, Poston G, Cunningham D, Ruers T, Primrose J. The clinical effectiveness and cost-effectiveness of ablative therapies in the management of liver metastases: systematic review and economic evaluation. Health Technol Assess. 2014;18:vii-viii, 1-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
107.  Ko S, Jo H, Yun S, Park E, Kim S, Seo HI. Comparative analysis of radiofrequency ablation and resection for resectable colorectal liver metastases. World J Gastroenterol. 2014;20:525-531.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 34]  [Cited by in F6Publishing: 31]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
108.  van Amerongen MJ, Jenniskens SFM, van den Boezem PB, Fütterer JJ, de Wilt JHW. Radiofrequency ablation compared to surgical resection for curative treatment of patients with colorectal liver metastases - a meta-analysis. HPB (Oxford). 2017;19:749-756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 77]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
109.  Petre EN, Sofocleous C. Thermal Ablation in the Management of Colorectal Cancer Patients with Oligometastatic Liver Disease. Visc Med. 2017;33:62-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
110.  Sotirchos VS, Petrovic LM, Gönen M, Klimstra DS, Do RK, Petre EN, Garcia AR, Barlas A, Erinjeri JP, Brown KT, Covey AM, Alago W, Brody LA, DeMatteo RP, Kemeny NE, Solomon SB, Manova-Todorova KO, Sofocleous CT. Colorectal Cancer Liver Metastases: Biopsy of the Ablation Zone and Margins Can Be Used to Predict Oncologic Outcome. Radiology. 2016;280:949-959.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 99]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
111.  Gurusamy K, Corrigan N, Croft J, Twiddy M, Morris S, Woodward N, Bandula S, Hochhauser D, Napp V, Pullan A, Jakowiw N, Prasad R, Damink SO, van Laarhoven CJHM, de Wilt JHW, Brown J, Davidson BR. Liver resection surgery versus thermal ablation for colorectal LiVer MetAstases (LAVA): study protocol for a randomised controlled trial. Trials. 2018;19:105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 37]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
112.  Isfordink CJ, Samim M, Braat MNGJA, Almalki AM, Hagendoorn J, Borel Rinkes IHM, Molenaar IQ. Portal vein ligation versus portal vein embolization for induction of hypertrophy of the future liver remnant: A systematic review and meta-analysis. Surg Oncol. 2017;26:257-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 46]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
113.  Shindoh J, Tzeng CW, Aloia TA, Curley SA, Zimmitti G, Wei SH, Huang SY, Gupta S, Wallace MJ, Vauthey JN. Portal vein embolization improves rate of resection of extensive colorectal liver metastases without worsening survival. Br J Surg. 2013;100:1777-1783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 80]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
114.  Al-Sharif E, Simoneau E, Hassanain M. Portal vein embolization effect on colorectal cancer liver metastasis progression: Lessons learned. World J Clin Oncol. 2015;6:142-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 14]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
115.  Giglio MC, Giakoustidis A, Draz A, Jawad ZAR, Pai M, Habib NA, Tait P, Frampton AE, Jiao LR. Oncological Outcomes of Major Liver Resection Following Portal Vein Embolization: A Systematic Review and Meta-analysis. Ann Surg Oncol. 2016;23:3709-3717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
116.  Lam VW, Laurence JM, Johnston E, Hollands MJ, Pleass HC, Richardson AJ. A systematic review of two-stage hepatectomy in patients with initially unresectable colorectal liver metastases. HPB (Oxford). 2013;15:483-491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 150]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
117.  Moris D, Ronnekleiv-Kelly S, Kostakis ID, Tsilimigras DI, Beal EW, Papalampros A, Dimitroulis D, Felekouras E, Pawlik TM. Operative Results and Oncologic Outcomes of Associating Liver Partition and Portal Vein Ligation for Staged Hepatectomy (ALPPS) Versus Two-Stage Hepatectomy (TSH) in Patients with Unresectable Colorectal Liver Metastases: A Systematic Review and Meta-Analysis. World J Surg. 2018;42:806-815.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 55]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
118.  Eshmuminov D, Raptis DA, Linecker M, Wirsching A, Lesurtel M, Clavien PA. Meta-analysis of associating liver partition with portal vein ligation and portal vein occlusion for two-stage hepatectomy. Br J Surg. 2016;103:1768-1782.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 105]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
119.  Sandström P, Røsok BI, Sparrelid E, Larsen PN, Larsson AL, Lindell G, Schultz NA, Bjørnbeth BA, Isaksson B, Rizell M, Björnsson B. ALPPS Improves Resectability Compared With Conventional Two-stage Hepatectomy in Patients With Advanced Colorectal Liver Metastasis: Results From a Scandinavian Multicenter Randomized Controlled Trial (LIGRO Trial). Ann Surg. 2018;267:833-840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 190]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
120.  Ulmer TF, de Jong C, Andert A, Bruners P, Heidenhain CM, Schoening W, Schmeding M, Neumann UP. ALPPS Procedure in Insufficient Hypertrophy After Portal Vein Embolization (PVE). World J Surg. 2017;41:250-257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
121.  Schadde E, Raptis DA, Schnitzbauer AA, Ardiles V, Tschuor C, Lesurtel M, Abdalla EK, Hernandez-Alejandro R, Jovine E, Machado M, Malago M, Robles-Campos R, Petrowsky H, Santibanes ED, Clavien PA. Prediction of Mortality After ALPPS Stage-1: An Analysis of 320 Patients From the International ALPPS Registry. Ann Surg. 2015;262:780-5; discussion 785-6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 168]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
122.  Hernandez-Alejandro R, Bertens KA, Pineda-Solis K, Croome KP. Can we improve the morbidity and mortality associated with the associating liver partition with portal vein ligation for staged hepatectomy (ALPPS) procedure in the management of colorectal liver metastases? Surgery. 2015;157:194-201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 90]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
123.  Björnsson B, Sparrelid E, Røsok B, Pomianowska E, Hasselgren K, Gasslander T, Bjørnbeth BA, Isaksson B, Sandström P. Associating liver partition and portal vein ligation for staged hepatectomy in patients with colorectal liver metastases--Intermediate oncological results. Eur J Surg Oncol. 2016;42:531-537.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 38]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
124.  Olthof PB, Huiskens J, Wicherts DA, Huespe PE, Ardiles V, Robles-Campos R, Adam R, Linecker M, Clavien PA, Koopman M, Verhoef C, Punt CJ, van Gulik TM, de Santibanes E. Survival after associating liver partition and portal vein ligation for staged hepatectomy (ALPPS) for advanced colorectal liver metastases: A case-matched comparison with palliative systemic therapy. Surgery. 2017;161:909-919.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
125.  Wanis KN, Ardiles V, Alvarez FA, Tun-Abraham ME, Linehan D, de Santibañes E, Hernandez-Alejandro R. Intermediate-term survival and quality of life outcomes in patients with advanced colorectal liver metastases undergoing associating liver partition and portal vein ligation for staged hepatectomy. Surgery. 2018;163:691-697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 34]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
126.  Karanicolas PJ, Jarnagin WR, Gonen M, Tuorto S, Allen PJ, DeMatteo RP, D'Angelica MI, Fong Y. Long-term outcomes following tumor ablation for treatment of bilateral colorectal liver metastases. JAMA Surg. 2013;148:597-601.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 71]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
127.  Faitot F, Faron M, Adam R, Elias D, Cimino M, Cherqui D, Vibert E, Castaing D, Cunha AS, Goéré D. Two-stage hepatectomy versus 1-stage resection combined with radiofrequency for bilobar colorectal metastases: a case-matched analysis of surgical and oncological outcomes. Ann Surg. 2014;260:822-7; discussion 827-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
128.  Evrard S, Poston G, Kissmeyer-Nielsen P, Diallo A, Desolneux G, Brouste V, Lalet C, Mortensen F, Stättner S, Fenwick S, Malik H, Konstantinidis I, DeMatteo R, D'Angelica M, Allen P, Jarnagin W, Mathoulin-Pelissier S, Fong Y. Combined ablation and resection (CARe) as an effective parenchymal sparing treatment for extensive colorectal liver metastases. PLoS One. 2014;9:e114404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 57]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
129.  André T, de Gramont A, Vernerey D, Chibaudel B, Bonnetain F, Tijeras-Raballand A, Scriva A, Hickish T, Tabernero J, Van Laethem JL, Banzi M, Maartense E, Shmueli E, Carlsson GU, Scheithauer W, Papamichael D, Möehler M, Landolfi S, Demetter P, Colote S, Tournigand C, Louvet C, Duval A, Fléjou JF, de Gramont A. Adjuvant Fluorouracil, Leucovorin, and Oxaliplatin in Stage II to III Colon Cancer: Updated 10-Year Survival and Outcomes According to BRAF Mutation and Mismatch Repair Status of the MOSAIC Study. J Clin Oncol. 2015;33:4176-4187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 366]  [Cited by in F6Publishing: 395]  [Article Influence: 43.9]  [Reference Citation Analysis (0)]
130.  Mitry E, Fields AL, Bleiberg H, Labianca R, Portier G, Tu D, Nitti D, Torri V, Elias D, O'Callaghan C, Langer B, Martignoni G, Bouché O, Lazorthes F, Van Cutsem E, Bedenne L, Moore MJ, Rougier P. Adjuvant chemotherapy after potentially curative resection of metastases from colorectal cancer: a pooled analysis of two randomized trials. J Clin Oncol. 2008;26:4906-4911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 407]  [Cited by in F6Publishing: 392]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
131.  Hasegawa K, Saiura A, Takayama T, Miyagawa S, Yamamoto J, Ijichi M, Teruya M, Yoshimi F, Kawasaki S, Koyama H, Oba M, Takahashi M, Mizunuma N, Matsuyama Y, Watanabe T, Makuuchi M, Kokudo N. Adjuvant Oral Uracil-Tegafur with Leucovorin for Colorectal Cancer Liver Metastases: A Randomized Controlled Trial. PLoS One. 2016;11:e0162400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 76]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
132.  Kanemitsu Y, Kato T, Shimizu Y, Inaba Y, Shimada Y, Nakamura K, Sato A, Moriya Y; Colorectal Cancer Study Group (CCSG) of Japan Clinical Oncology Group. A randomized phase II/III trial comparing hepatectomy followed by mFOLFOX6 with hepatectomy alone as treatment for liver metastasis from colorectal cancer: Japan Clinical Oncology Group Study JCOG0603. Jpn J Clin Oncol. 2009;39:406-409.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 47]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
133.  Ychou M, Hohenberger W, Thezenas S, Navarro M, Maurel J, Bokemeyer C, Shacham-Shmueli E, Rivera F, Kwok-Keung Choi C, Santoro A. A randomized phase III study comparing adjuvant 5-fluorouracil/folinic acid with FOLFIRI in patients following complete resection of liver metastases from colorectal cancer. Ann Oncol. 2009;20:1964-1970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 184]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
134.  Kemeny NE, Jarnagin WR, Capanu M, Fong Y, Gewirtz AN, Dematteo RP, D'Angelica MI. Randomized phase II trial of adjuvant hepatic arterial infusion and systemic chemotherapy with or without bevacizumab in patients with resected hepatic metastases from colorectal cancer. J Clin Oncol. 2011;29:884-889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 87]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
135.  Turan N, Benekli M, Koca D, Ustaalioglu BO, Dane F, Ozdemir N, Ulas A, Oztop I, Gumus M, Ozturk MA, Berk V, Kucukoner M, Uner A, Balakan O, Helvaci K, Ozkan S, Yilmaz U, Buyukberber S; Anatolian Society of Medical Oncology. Adjuvant systemic chemotherapy with or without bevacizumab in patients with resected liver metastases from colorectal cancer. Oncology. 2013;84:14-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
136.  Snoeren N, Voest EE, Bergman AM, Dalesio O, Verheul HM, Tollenaar RA, van der Sijp JR, Schouten SB, Rinkes IH, van Hillegersberg R. A randomized two arm phase III study in patients post radical resection of liver metastases of colorectal cancer to investigate bevacizumab in combination with capecitabine plus oxaliplatin (CAPOX) vs CAPOX alone as adjuvant treatment. BMC Cancer. 2010;10:545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 22]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
137.  Kemeny NE, Gonen M. Hepatic arterial infusion after liver resection. N Engl J Med. 2005;352:734-735.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 167]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
138.  Groot Koerkamp B, Sadot E, Kemeny NE, Gönen M, Leal JN, Allen PJ, Cercek A, DeMatteo RP, Kingham TP, Jarnagin WR, D'Angelica MI. Perioperative Hepatic Arterial Infusion Pump Chemotherapy Is Associated With Longer Survival After Resection of Colorectal Liver Metastases: A Propensity Score Analysis. J Clin Oncol. 2017;35:1938-1944.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 89]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
139.  Kemeny NE, Chou JF, Boucher TM, Capanu M, DeMatteo RP, Jarnagin WR, Allen PJ, Fong YC, Cercek A, D'Angelica MI. Updated long-term survival for patients with metastatic colorectal cancer treated with liver resection followed by hepatic arterial infusion and systemic chemotherapy. J Surg Oncol. 2016;113:477-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 58]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
140.  Goéré D, Pignon JP, Gelli M, Elias D, Benhaim L, Deschamps F, Caramella C, Boige V, Ducreux M, de Baere T, Malka D. Postoperative hepatic arterial chemotherapy in high-risk patients as adjuvant treatment after resection of colorectal liver metastases - a randomized phase II/III trial - PACHA-01 (NCT02494973). BMC Cancer. 2018;18:787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
141.  Massmann A, Rodt T, Marquardt S, Seidel R, Thomas K, Wacker F, Richter GM, Kauczor HU, Bücker A, Pereira PL, Sommer CM. Transarterial chemoembolization (TACE) for colorectal liver metastases--current status and critical review. Langenbecks Arch Surg. 2015;400:641-659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
142.  Foubert F, Matysiak-Budnik T, Touchefeu Y. Options for metastatic colorectal cancer beyond the second line of treatment. Dig Liver Dis. 2014;46:105-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
143.  Lam VW, Spiro C, Laurence JM, Johnston E, Hollands MJ, Pleass HC, Richardson AJ. A systematic review of clinical response and survival outcomes of downsizing systemic chemotherapy and rescue liver surgery in patients with initially unresectable colorectal liver metastases. Ann Surg Oncol. 2012;19:1292-1301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 128]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
144.  Kanat O. Current treatment options for patients with initially unresectable isolated colorectal liver metastases. World J Clin Oncol. 2016;7:9-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
145.  Falcone A, Ricci S, Brunetti I, Pfanner E, Allegrini G, Barbara C, Crinò L, Benedetti G, Evangelista W, Fanchini L, Cortesi E, Picone V, Vitello S, Chiara S, Granetto C, Porcile G, Fioretto L, Orlandini C, Andreuccetti M, Masi G; Gruppo Oncologico Nord Ovest. Phase III trial of infusional fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI) compared with infusional fluorouracil, leucovorin, and irinotecan (FOLFIRI) as first-line treatment for metastatic colorectal cancer: the Gruppo Oncologico Nord Ovest. J Clin Oncol. 2007;25:1670-1676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 830]  [Cited by in F6Publishing: 838]  [Article Influence: 49.3]  [Reference Citation Analysis (0)]
146.  Saltz LB, Clarke S, Díaz-Rubio E, Scheithauer W, Figer A, Wong R, Koski S, Lichinitser M, Yang TS, Rivera F, Couture F, Sirzén F, Cassidy J. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol. 2008;26:2013-2019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2147]  [Cited by in F6Publishing: 2204]  [Article Influence: 137.8]  [Reference Citation Analysis (0)]
147.  Tomasello G, Petrelli F, Ghidini M, Russo A, Passalacqua R, Barni S. FOLFOXIRI Plus Bevacizumab as Conversion Therapy for Patients With Initially Unresectable Metastatic Colorectal Cancer: A Systematic Review and Pooled Analysis. JAMA Oncol. 2017;3:e170278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 126]  [Article Influence: 18.0]  [Reference Citation Analysis (1)]
148.  Ye LC, Liu TS, Ren L, Wei Y, Zhu DX, Zai SY, Ye QH, Yu Y, Xu B, Qin XY, Xu J. Randomized controlled trial of cetuximab plus chemotherapy for patients with KRAS wild-type unresectable colorectal liver-limited metastases. J Clin Oncol. 2013;31:1931-1938.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 293]  [Cited by in F6Publishing: 297]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
149.  Petrelli F, Barni S; Anti-EGFR agents for liver metastases. Resectability and outcome with anti-EGFR agents in patients with KRAS wild-type colorectal liver-limited metastases: a meta-analysis. Int J Colorectal Dis. 2012;27:997-1004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 48]  [Article Influence: 4.0]  [Reference Citation Analysis (1)]
150.  Peeters M, Tabernero J, Douillard JY, Siena S, Davison C, Braun S, Sidhu R, Öhrling K. Resection rates and survival in patients with wild-type KRAS/NRAS metastatic colorectal cancer and liver metastases: data from the PRIME study. In: Eggermont AMM, editors. Abstract book for Markers in cancer: a joint meeting by ASCO, EORTC and NCI; 2013 Nov 7-9. Brussels, Belgium. Eur J Cancer. 2013;49 suppl 4:S17-18.  [PubMed]  [DOI]  [Cited in This Article: ]
151.  DʼAngelica MI, Correa-Gallego C, Paty PB, Cercek A, Gewirtz AN, Chou JF, Capanu M, Kingham TP, Fong Y, DeMatteo RP, Allen PJ, Jarnagin WR, Kemeny N. Phase II trial of hepatic artery infusional and systemic chemotherapy for patients with unresectable hepatic metastases from colorectal cancer: conversion to resection and long-term outcomes. Ann Surg. 2015;261:353-360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 129]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
152.  Chapelle N, Matysiak-Budnik T, Douane F, Metairie S, Rougier P, Touchefeu Y. Hepatic arterial infusion in the management of colorectal cancer liver metastasis: Current and future perspectives. Dig Liver Dis. 2018;50:220-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
153.  Lévi FA, Boige V, Hebbar M, Smith D, Lepère C, Focan C, Karaboué A, Guimbaud R, Carvalho C, Tumolo S, Innominato P, Ajavon Y, Truant S, Castaing D, De Baere T, Kunstlinger F, Bouchahda M, Afshar M, Rougier P, Adam R, Ducreux M; Association Internationale pour Recherche sur Temps Biologique et Chronothérapie (ARTBC International). Conversion to resection of liver metastases from colorectal cancer with hepatic artery infusion of combined chemotherapy and systemic cetuximab in multicenter trial OPTILIV. Ann Oncol. 2016;27:267-274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 74]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
154.  Cauchy F, Aussilhou B, Dokmak S, Fuks D, Gaujoux S, Farges O, Faivre S, Lepillé D, Belghiti J. Reappraisal of the risks and benefits of major liver resection in patients with initially unresectable colorectal liver metastases. Ann Surg. 2012;256:746-52; discussion 752-4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
155.  Gillams A, Goldberg N, Ahmed M, Bale R, Breen D, Callstrom M, Chen MH, Choi BI, de Baere T, Dupuy D, Gangi A, Gervais D, Helmberger T, Jung EM, Lee F, Lencioni R, Liang P, Livraghi T, Lu D, Meloni F, Pereira P, Piscaglia F, Rhim H, Salem R, Sofocleous C, Solomon SB, Soulen M, Tanaka M, Vogl T, Wood B, Solbiati L. Thermal ablation of colorectal liver metastases: a position paper by an international panel of ablation experts, The Interventional Oncology Sans Frontières meeting 2013. Eur Radiol. 2015;25:3438-3454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 195]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
156.  Shady W, Petre EN, Gonen M, Erinjeri JP, Brown KT, Covey AM, Alago W, Durack JC, Maybody M, Brody LA, Siegelbaum RH, D'Angelica MI, Jarnagin WR, Solomon SB, Kemeny NE, Sofocleous CT. Percutaneous Radiofrequency Ablation of Colorectal Cancer Liver Metastases: Factors Affecting Outcomes--A 10-year Experience at a Single Center. Radiology. 2016;278:601-611.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 240]  [Article Influence: 26.7]  [Reference Citation Analysis (0)]
157.  Ruers T, Punt C, Van Coevorden F, Pierie JP, Borel-Rinkes I, Ledermann JA, Poston G, Bechstein W, Lentz MA, Mauer M, Van Cutsem E, Lutz MP, Nordlinger B; EORTC Gastro-Intestinal Tract Cancer Group, Arbeitsgruppe Lebermetastasen und—tumoren in der Chirurgischen Arbeitsgemeinschaft Onkologie (ALM-CAO) and the National Cancer Research Institute Colorectal Clinical Study Group (NCRI CCSG). Radiofrequency ablation combined with systemic treatment versus systemic treatment alone in patients with non-resectable colorectal liver metastases: a randomized EORTC Intergroup phase II study (EORTC 40004). Ann Oncol. 2012;23:2619-2626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 272]  [Cited by in F6Publishing: 284]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
158.  Ruers T, Punt CJA, van Coevorden F, Pierie JP, Borel Rinkes I, Ledermann JA, Poston GJ, Bechstein WO, Lentz M, Mauer ME, van Cutsem E, Lutz MP, Nordlinger B. Radiofrequency ablation (RFA) combined with chemotherapy for unresectable colorectal liver metastases (CRC LM): long-term survival results of a randomized phase II study of the EORTC-NCRI CCSG-ALM Intergroup 40004 (CLOCC). ASCO Annual Meeting 2015. J Clin Oncol. 2015;33:abstr 3501.  [PubMed]  [DOI]  [Cited in This Article: ]
159.  Correa-Gallego C, Fong Y, Gonen M, D'Angelica MI, Allen PJ, DeMatteo RP, Jarnagin WR, Kingham TP. A retrospective comparison of microwave ablation vs. radiofrequency ablation for colorectal cancer hepatic metastases. Ann Surg Oncol. 2014;21:4278-4283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 122]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
160.  Huo YR, Eslick GD. Microwave Ablation Compared to Radiofrequency Ablation for Hepatic Lesions: A Meta-Analysis. J Vasc Interv Radiol. 2015;26:1139-1146.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 65]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
161.  Hosein PJ, Echenique A, Loaiza-Bonilla A, Froud T, Barbery K, Rocha Lima CM, Yrizarry JM, Narayanan G. Percutaneous irreversible electroporation for the treatment of colorectal cancer liver metastases with a proposal for a new response evaluation system. J Vasc Interv Radiol. 2014;25:1233-1239.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 42]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
162.  Vogl TJ, Gruber T, Balzer JO, Eichler K, Hammerstingl R, Zangos S. Repeated transarterial chemoembolization in the treatment of liver metastases of colorectal cancer: prospective study. Radiology. 2009;250:281-289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 108]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
163.  Albert M, Kiefer MV, Sun W, Haller D, Fraker DL, Tuite CM, Stavropoulos SW, Mondschein JI, Soulen MC. Chemoembolization of colorectal liver metastases with cisplatin, doxorubicin, mitomycin C, ethiodol, and polyvinyl alcohol. Cancer. 2011;117:343-352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 104]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
164.  Xing M, Kooby DA, El-Rayes BF, Kokabi N, Camacho JC, Kim HS. Locoregional therapies for metastatic colorectal carcinoma to the liver--an evidence-based review. J Surg Oncol. 2014;110:182-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
165.  Aliberti C, Fiorentini G, Muzzio PC, Pomerri F, Tilli M, Dallara S, Benea G. Trans-arterial chemoembolization of metastatic colorectal carcinoma to the liver adopting DC Bead®, drug-eluting bead loaded with irinotecan: results of a phase II clinical study. Anticancer Res. 2011;31:4581-4587.  [PubMed]  [DOI]  [Cited in This Article: ]
166.  Fiorentini G, Aliberti C, Tilli M, Mulazzani L, Graziano F, Giordani P, Mambrini A, Montagnani F, Alessandroni P, Catalano V, Coschiera P. Intra-arterial infusion of irinotecan-loaded drug-eluting beads (DEBIRI) versus intravenous therapy (FOLFIRI) for hepatic metastases from colorectal cancer: final results of a phase III study. Anticancer Res. 2012;32:1387-1395.  [PubMed]  [DOI]  [Cited in This Article: ]
167.  Akinwande O, Miller A, Hayes D, O'Hara R, Tomalty D, Martin RC. Concomitant capecitabine with hepatic delivery of drug eluting beads in metastatic colorectal cancer. Anticancer Res. 2014;34:7239-7245.  [PubMed]  [DOI]  [Cited in This Article: ]
168.  Martin RC 2nd, Scoggins CR, Schreeder M, Rilling WS, Laing CJ, Tatum CM, Kelly LR, Garcia-Monaco RD, Sharma VR, Crocenzi TS, Strasberg SM. Randomized controlled trial of irinotecan drug-eluting beads with simultaneous FOLFOX and bevacizumab for patients with unresectable colorectal liver-limited metastasis. Cancer. 2015;121:3649-3658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 103]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
169.  Yamakado K, Inaba Y, Sato Y, Yasumoto T, Hayashi S, Yamanaka T, Nobata K, Takaki H, Nakatsuka A. Radiofrequency Ablation Combined with Hepatic Arterial Chemoembolization Using Degradable Starch Microsphere Mixed with Mitomycin C for the Treatment of Liver Metastasis from Colorectal Cancer: A Prospective Multicenter Study. Cardiovasc Intervent Radiol. 2017;40:560-567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
170.  Hendlisz A, Van den Eynde M, Peeters M, Maleux G, Lambert B, Vannoote J, De Keukeleire K, Verslype C, Defreyne L, Van Cutsem E, Delatte P, Delaunoit T, Personeni N, Paesmans M, Van Laethem JL, Flamen P. Phase III trial comparing protracted intravenous fluorouracil infusion alone or with yttrium-90 resin microspheres radioembolization for liver-limited metastatic colorectal cancer refractory to standard chemotherapy. J Clin Oncol. 2010;28:3687-3694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 293]  [Cited by in F6Publishing: 258]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
171.  Wasan HS, Gibbs P, Sharma NK, Taieb J, Heinemann V, Ricke J, Peeters M, Findlay M, Weaver A, Mills J, Wilson C, Adams R, Francis A, Moschandreas J, Virdee PS, Dutton P, Love S, Gebski V, Gray A; FOXFIRE trial investigators; SIRFLOX trial investigators; FOXFIRE-Global trial investigators, van Hazel G, Sharma RA. First-line selective internal radiotherapy plus chemotherapy versus chemotherapy alone in patients with liver metastases from colorectal cancer (FOXFIRE, SIRFLOX, and FOXFIRE-Global): a combined analysis of three multicentre, randomised, phase 3 trials. Lancet Oncol. 2017;18:1159-1171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 211]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
172.  Mocellin S, Pilati P, Lise M, Nitti D. Meta-analysis of hepatic arterial infusion for unresectable liver metastases from colorectal cancer: the end of an era? J Clin Oncol. 2007;25:5649-5654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 141]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
173.  Levy J, Zuckerman J, Garfinkle R, Acuna SA, Touchette J, Vanounou T, Pelletier JS. Intra-arterial therapies for unresectable and chemorefractory colorectal cancer liver metastases: a systematic review and meta-analysis. HPB (Oxford). 2018;20:905-915.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
174.  Elias D, Viganò L, Orsi F, Scorsetti M, Comito T, Lerut J, Cosola D, Torzilli G. New Perspectives in the Treatment of Colorectal Metastases. Liver Cancer. 2016;6:90-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
175.  Petrelli F, Comito T, Barni S, Pancera G, Scorsetti M, Ghidini A; SBRT for CRC liver metastases. Stereotactic body radiotherapy for colorectal cancer liver metastases: A systematic review. Radiother Oncol. 2018;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 83]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
176.  Hagness M, Foss A, Line PD, Scholz T, Jørgensen PF, Fosby B, Boberg KM, Mathisen O, Gladhaug IP, Egge TS, Solberg S, Hausken J, Dueland S. Liver transplantation for nonresectable liver metastases from colorectal cancer. Ann Surg. 2013;257:800-806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 225]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
177.  Dueland S, Guren TK, Hagness M, Glimelius B, Line PD, Pfeiffer P, Foss A, Tveit KM. Chemotherapy or liver transplantation for nonresectable liver metastases from colorectal cancer? Ann Surg. 2015;261:956-960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 114]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
178.  Toso C, Pinto Marques H, Andres A, Castro Sousa F, Adam R, Kalil A, Clavien PA, Furtado E, Barroso E, Bismuth H; Compagnons Hépato-Biliaires Group. Liver transplantation for colorectal liver metastasis: Survival without recurrence can be achieved. Liver Transpl. 2017;23:1073-1076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 78]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
179.  Sapisochin G. Assessment of a protocol using a combination of neo-adjuvant chemotherapy plus living donor liver transplantation for non-resectable liver metastases from colorectal cancer. [accessed. 2018;ClinicalTrials.gov [Internet]. Bethesda (MD): U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/ct2/show/NCT02864485 Clinical Trials.gov Identifier: NCT02864485.  [PubMed]  [DOI]  [Cited in This Article: ]
180.  Dueland S. A randomized controlled clinical trial to evaluate the benefit and efficacy of liver transplantation as treatment for selected patients with liver metastases from colorectal carcinoma. [accessed. 2018;ClinicalTrials.gov [Internet]. Bethesda (MD): U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/ct2/show/NCT01479608 ClinicalTrials.gov Identifier: NCT01479608.  [PubMed]  [DOI]  [Cited in This Article: ]
181.  Dueland S. A Phase I/II clinical trial to evaluate the benefit and efficacy of liver resection and partial liver segment 2/3 transplantation with delayed total hepatectomy as treatment for selected patients with liver metastases from colorectal carcinoma. [accessed. 2018;ClinicalTrials.gov [Internet]. Bethesda (MD): U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/ct2/show/NCT02215889 ClinicalTrials.gov Identifier: NCT02215889.  [PubMed]  [DOI]  [Cited in This Article: ]
182.  Adam R. Curative potential of liver transplantation in patients with definitively unresectable colorectal liver metastases (CLM) treated by chemotherapy: a prospective multicentric randomized trial. [accessed. 2018;ClinicalTrials.gov [Internet]. Bethesda (MD): U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/ct2/show/NCT02597348 ClinicalTrials.gov Identifier: NCT02597348.  [PubMed]  [DOI]  [Cited in This Article: ]