Skip to main content
ecancermedicalscience logoLink to ecancermedicalscience
. 2017 Jun 29;11:750. doi: 10.3332/ecancer.2017.750

Intraoperative radiotherapy: review of techniques and results

Avinash Pilar 1, Meetakshi Gupta 1, Sarbani Ghosh Laskar 1, Siddhartha Laskar 1
PMCID: PMC5493441  PMID: 28717396

Abstract

Intraoperative radiotherapy (IORT) is a technique that involves precise delivery of a large dose of ionising radiation to the tumour or tumour bed during surgery. Direct visualisation of the tumour bed and ability to space out the normal tissues from the tumour bed allows maximisation of the dose to the tumour while minimising the dose to normal tissues. This results in an improved therapeutic ratio with IORT. Although it was introduced in the 1960s, it has seen a resurgence of popularity with the introduction of self-shielding mobile linear accelerators and low-kV IORT devices, which by eliminating the logistical issues of transport of the patient during surgery for radiotherapy or building a shielded operating room, has enabled its wider use in the community.

Electrons, low-kV X-rays and HDR brachytherapy are all different methods of IORT in current clinical use. Each method has its own unique set of advantages and disadvantages, its own set of indications where one may be better suited than the other, and each requires a specific kind of expertise.

IORT has demonstrated its efficacy in a wide variety of intra-abdominal tumours, recurrent colorectal cancers, recurrent gynaecological cancers, and soft-tissue tumours. Recently, it has emerged as an attractive treatment option for selected, early-stage breast cancer, owing to the ability to complete the entire course of radiotherapy during surgery. IORT has been used in a multitude of roles across these sites, for dose escalation (retroperitoneal sarcoma), EBRT dose de-escalation (paediatric tumours), as sole radiation modality (early breast cancers) and as a re-irradiation modality (recurrent rectal and gynaecological cancers).

This article aims to provide a review of the rationale, techniques, and outcomes for IORT across different sites relevant to current clinical practice.

Keywords: IORT, techniques, indications, outcomes, complications

Background

Intraoperative radiation therapy (IORT) constitutes delivery of radiation to the tumour/tumour bed while the area is exposed during surgery. IORT is capable of delivering high doses of radiation, precisely to the tumour bed with minimal exposure to the surrounding healthy tissues.

Abe et al. from the University of Kyoto, Japan, were the first to introduce IORT in the early 1960s reporting its use in various intra-abdominal tumours [13].

IORT is typically used in combination with other modalities like maximal surgical resection, external beam radiotherapy (EBRT) or chemotherapy as a part of the multidisciplinary approach.

Efficacy of IORT has been reported in a wide variety of sites like locally advanced and recurrent rectal cancer, retroperitoneal sarcoma, pancreatic cancer, early breast cancer, and selected gynaecologic and genitourinary malignancies.

Rationale for the use of IORT

Traditionally, surgery is followed by EBRT in most solid tumours for the elimination of any microscopic residual disease and reducing the risk of local recurrence. However, EBRT in the post-operative setting has the following drawbacks:

  • The usual delay between the surgical removal of the tumour and EBRT may allow repopulation of the tumour cells.

  • Difficulty in tumour bed localisation or use of larger margins, which may increase normal tissue morbidity.

Most solid tumours exhibit a dose–response relationship, the likelihood of local control improving with increasing dose; however, there are limitations to the doses that can be delivered even with conformal EBRT techniques due to the presence of dose-limiting structures adjacent to the tumour/tumour bed. Especially, in the setting of gross residual disease, doses with EBRT may never be sufficient to achieve adequate local control without causing significant morbidity.

IORT allows

  • Precise localisation of the tumour bed and targeted delivery of high-dose radiation to the tumour bed.

  • Minimal exposure of the dose-limiting normal tissues that are displaced away from the tumour bed and shielded from radiation.

  • Opportunities for dose escalation beyond that which can be achieved with EBRT.

  • Opportunities for re-irradiation especially in recurrent cancers where further irradiation with EBRT may not be possible.

Thus, IORT can deliver higher total effective dose to the tumour bed, facilitate dose escalation without significantly increasing normal tissue complications and improve therapeutic ratio compared with EBRT.

IORT may be used alone or in combination with conventionally fractionated EBRT. Most centres use it in combination with EBRT, as it seems to provide the best therapeutic ratio (decreased risk of late normal tissue damage due to the use of fractionation for some part of the dose).

Methods of IORT

Several methods have been used to deliver IORT. Electron beams (electron IORT/IOERT), X-rays (kV IORT) and High-dose-rate brachytherapy (HDR IORT) are some of the commonly used methods for the delivery of IORT in current clinical practice.

Electron IORT

Introduction of electron IORT (IOERT) marked the beginning of the IORT era in the early 1960s [3, 4]. Using variable electron energies depth dose distribution could be controlled to provide uniform dose to target area. However, patients needed to be transported from the operating room (O.R) to the radiation department during surgery, posing logistical issues related to transportation and sterilisation [2, 5]. These problems were overcome with the use of dedicated IOERT facilities, which were quite expensive because of added costs of shielding the O.R and dedicated linear accelerator requirements, limiting their use to few centres in the United States and Europe. The advent of miniaturised, self-shielded, mobile linear accelerators [6] (Novac7, Hitesys SPA, Aprillia, Italy; 7–10 MeV and the Mobetron, IntraOp Medical Corporation, Sunnyvale, CA, USA; 4–12 MeV) in the 1990s, has brought about resurgence of IORT and allowed its use in many centres across the world while reducing the costs. Greater depth of penetration and dose homogeneity relative to HDR-IORT or kV IORT is possible with these devices. They come with applicators of different shapes and sizes, for the treatment of various sites and can deliver the treatment in a matter of minutes [7]. However, these applicators are rigid, thus challenging to use in difficult sites (pelvis and narrow cavities) and can treat a maximum diameter of 15 cm only, larger volumes requiring multiple, closely placed fields. Abutment of fields to treat a wider area is made possible by the use of rectangular applicators or D-shaped applicators called ‘Squircle’.

HDR IORT

HDR brachytherapy offers distinct dosimetric advantages due to its steep dose fall off and has the ability to deliver high doses to the tumour bed while reducing doses to nearby critical structures, these characteristics of HDR brachytherapy make it well suited for the purpose of IORT. Since many centres already own a HDR after loading machine, which can be transported to the OR for IORT, it reduces the cost of dedicated system; however, like IOERT, a shielded O.R or a shielded room in the O.R complex becomes necessary for HDR IORT. HDR IORT in most centres is delivered using surface applicators like Harrison–Anderson–Mick (HAM) applicator [8, 9] or superflab [10, 11] applicators and prescribed at 0.5–1 cm depth. These applicators are flexible, can treat relatively uneven surfaces and come in larger sizes for larger surfaces. Disadvantages of HDR IORT are reduced depth of penetration and prolonged treatment time relative to IOERT.

KV IORT

With increasing use of IOERT in the 1980s, orthovoltage X-rays were attempted for use in IORT to reduce the shielding costs of the OR. However, poor uniformity, higher bone doses and prolonged treatment time quickly reduced the interest in their use. Recently, low-kV (20–50 kV) mobile IORT devices like Intrabeam, (Carl Zeiss AG, Germany) and Axxent Electronic Brachytherapy System (Xoft Inc., Fremont, California) are gaining popularity for use in IORT. They have steep dose gradients and do not require special shielding requirements. They come with spherical applicators and have a very limited depth of penetration of 0.5–1 cm. They are therefore best suited for spherically shaped target volumes as in breast cancer.

With a strong oncological rationale at its heart, IORT in its various forms has been tested throughout the evolution of radiotherapy and has weathered the tests of time and technology showing periodic resurgences with the advent of newer technology. The following section will focus on recently published results to describe the current role of IORT across various sites.

Search strategy and selection criteria

A literature search was performed through the PubMed database by using the following terms: ‘intraoperative radiotherapy/IORT’, ‘head and neck cancer’, ‘breast cancer’, ‘colorectal/rectal/colon cancers’, ‘pancreas/pancreatic cancers’, ‘gastric/stomach cancer’, ‘soft-tissue sarcoma/sarcoma’, ‘paediatric/childhood cancers’, ‘gynaecological cancer’, ‘uterine/endometrial cancer’, ‘cervical/cervix cancer’, ‘renal/kidney cancer’, “bladder cancer”, and “prostate cancer”. IORT was defined as single large dose delivered intraoperatively during surgery, articles of perioperative brachytherapy with continuous low-dose rate or pulsed dose rate or HDR with multiple small fractions, delivered over subsequent days post-surgery were not included in this review. Search was limited to articles published between 1995 and 2017. Reviews, case reports and data presented, only as an abstract at conferences were excluded. Whenever updated data from the same institute was available, earlier articles with smaller numbers were not included. For the purpose of uniformity, in the respective sections, reports combining the results of primary with recurrent colorectal cancers, extremity sarcomas with retroperitoneal sarcomas and metastatic pancreatic cancers with locally advanced pancreatic cancers together were not included in the review. A total of 123 articles were finally included in the review.

Clinical results with IORT

Head and neck cancers

Despite the use of multidisciplinary treatment protocols locoregional recurrences occur in more than 30% of locoregionally advanced head and neck cancers [1215]. Outcomes are poor even after surgical salvage with high rates of local failure. Re-irradiation, in this setting, has shown to improve local control [16]. However, persistent late sequelae from previous course of radiotherapy (RT) may hamper the chances of effective re-irradiation with EBRT. IORT is an attractive tool in this setting.

Many retrospective series [1722] have demonstrated the efficacy of IORT in recurrent head and neck cancer after gross total resection (Table 1). Both IOERT and HDR IORT have been used to deliver IORT in recurrent head and neck cancer. Patients selected for IORT mainly consisted of recurrent or persistent cancers, who have been previously irradiated and delivery of sufficient doses of EBRT was not possible at the time of recurrence. Most studies have shown effective local control with acceptable complications [17, 1922]. Resection status at salvage was the most important factor determining local control [17, 19, 21]. Microscopically residual tumours did better with IORT [23], gross residual disease however did not [20, 23]. Adjuvant EBRT after IORT appears to further improve local control, however the small sample size of these studies precludes any definite conclusions [20, 23]. Wound complications, osteoradionecrosis (ORN), fistulae, and neuropathy are the most common complications [1722] after IOERT; however, these are rare with doses less than 20 Gy [22] and no different than that of re-irradiation with EBRT [16]. Carotid artery blow out is a rare but a fatal complication that may occur after IORT. Attempts should be made whenever possible to shield or space out the major vessels and nerves from the treatment field.

Table 1. Studies of IORT in recurrent head and neck cancer after gross total resection.

Author/Year Sample size Study design IORT type IORT dose (Gy) Prior RT (%) Adj. RT (%) Median follow-up LC (%) OS (%) Toxicity grade 3 or >
Scala et al 2013 [20] 76 (100% recurrent) Retrospective HDR IORT 10–17.5 71 24 11 62 (2yr) 42 (2yr) Total -6%
Neuropathy-1*
Wound-1*
Ziedan et al 2012 [21] 96 (48% recurrent) Retrospective (parotid cancers) IOERT 15–20 55 57 67 68.5 (3yr) 66.1 (3yr) Total -27%
Fistula-4*
ORN-4*
Neuropathy-1*
Ziedan et al 2011 [22] 231 (89% recurrent) Retrospective IOERT 15–20 81 21 12 55 (3yr) 34 (3yr) Total-27%
ORN-8*
Neuropathy-8*
Fistulas-20*
Perry et al 2010 [19] 34(100% recurrent) Retrospective HDR IORT 10–20 100 15 23 56 (2yr) 55 (2yr) Total-29%
Wound-3*
ORN-1*
Neuropathy-1*
Chen et al 2007 [17] 137 (100% recurrent) Retrospective IOERT 10–18 83 26 41 61 (3yr) 36 (3yr) Total-6%
Wound-4*
Fistula-2*
Neuropathy-1*
Pinheiro et al 2002 [23] 34- SCC 10- non-SCC Retrospective IOERT 12.5–22.5 64 36 75.6 for living
patients
46 52 (2yr) 32 50 (2yr) Total-7%
Fistula-1*
Neuropathy-1*
Carotid blowout-1*
Nag et al 1998 [18] 38 (100% recurrent) Retrospective IOERT 15–20 100 0 30 19 (1yr) 21% (1yr) Total -16%
Fistula-2*

Adj. RT: Adjuvant post-IORT RT, LC: local control, OS: overall Survival,

*

Number of patients.

Breast cancer

The majority of breast cancer recurrences after breast conservation surgery and whole breast irradiation (WBI) occur in the tumour bed, questioning the need for WBI. This has led to widespread adoption of accelerated partial breast irradiation (APBI) in women with early breast cancer without adverse features. IORT has seen a growing interest in early breast cancer as a modality of delivering APBI in a single fraction.

Several phase-II trials [24, 25] and prospective series [6, 26] have shown excellent early tumour control, survival, and cosmetic outcomes. Two large phase-III studies TARGIT-A (targeted intraoperative radiotherapy) [27] and ELIOT (intraoperative radiotherapy with electrons) [28], have evaluated the role of IORT as single-dose, partial breast irradiation treatment compared to standard, conventionally fractionated WBI for highly selected patients with relatively low-risk early-stage invasive breast cancer.

Table 2 summarises the relevant differences in the two trials with the 5-year results. Both the Eliot and TARGIT trials demonstrated significantly higher recurrence rates compared to WBI; however, the results were reported to be within the predefined statistical margin for equivalence/non-inferiority. Also, in both the trials, fewer skin side effects were seen in the IORT group compared to those in the WBI group.

Table 2. Randomised control trials of IORT versus WBI in early-breast cancer.

Trial name Sample size Age Inclusion criteria IORT type IORT dose Trial design Local recurrence (5yr) Overall survival/mortality (5yr)
ELIOT [28] 1305 median follow-up: 5.8 years > 48 years any invasive cancer < 2.5 cm IOERT ELIOT:21 Gy/1# to tumour bed with 6–9 MeV electrons
WBI:50 Gy/25# + 10 Gy/5# boost
Equivalence trial:
Statistical margin was local recurrence of 7.5% in the IORT group
ELIOT:4.4% (95% C.I: 2.7–6.1)
WBI: 0.4%
p < 0.0001
ELIOT:96.8%
WBI: 96.9%
TARGIT-A [27]
3451 Median follow-up:
2.5 years
> 45years T1-2, N0, 1 IDC < 3.5 cm
(If EIC or ILC on final histology,
add whole breast RT)
X-ray IORT (50 kv X-ray) TARGIT: 20 Gy to tumour bed
5–7 Gy at 1 cm depth
WBI: 40–56 Gy with or without boost 10–16 Gy
Non-inferiority trial:
Statistical margin was 2.5% difference in local recurrence at 5 years
TARGIT:3.3% (95% CI: 2.1–5.1)
WBI:1.3%
(95% CI: 0.7–2.5)
p = 0.042
TARGIT:3.9%
WBI: 5.3
p = 0.009

EIC: extensive intraductal component, ILC: invasive lobular cancer, WBI: whole breast RT.

TARGIT-A trial also reported significantly lower non-breast cancer deaths in the TARGIT group (p = 0.0086). This difference was attributed to fewer radiotherapy-related cardiovascular deaths in the TARGIT group; however, radiotherapy-related cardiovascular deaths may not become apparent so early in the follow-up period and these differences could have resulted due to imbalance in the treatment arms [2932]. The TARGIT-A trial has also come in for criticism related to its statistical assumptions [3335]. Though the trial seems to show a non-inferiority in 5-year local recurrence rates, median follow-up of all randomised patients is just 29 months which is too early to make assumptions regarding local recurrence rate at 5 years and also the authors seem to have misinterpreted the non-inferiority criterion, which require the upper confidence interval (CI) be less than the predefined non inferiority level of 2.5% [3335].

To summarise the trials of single-dose IORT, both ELIOT trial (IOERT) and TARGIT trial (kV-IORT) demonstrated a higher recurrence rate compared to WBI, although within the equivalence margin [36]. TARGIT-A (KV-IORT) also requires a longer follow-up before drawing definite conclusions and adopting it for widespread use in place of WBI [35, 36]. It is prudent to use these techniques in a highly selected group of low-risk early-breast cancer to achieve acceptable results. Leonardi et al [37, 38] used the American Society for Therapeutic Radiation Oncology (ASTRO) consensus statement [39] and the Groupe Européen de Curiethérapie–European Society for Therapeutic Radiology and Oncology (GEC–ESTRO) recommendations [40] for APBI patient selection, to stratify 1822 patients treated with ELIOT outside the trial into different risk groups, 16% of women met ASTRO suitable criteria and 31% were good candidates as per GEC-ESTRO recommendations, local recurrence rates were 1.5% and 1.9%, respectively. Thirty-six per cent of women who had favourable biology disease with a luminal-A subtype also showed a very low local recurrence rate of 1.7% irrespective of the risk group. Therefore, ASTRO suitable, GEC ESTRO good and luminal-A subtype identify a subset of women, who may be safely treated with single-dose IORT with acceptable results [3638, 41].

Though it may not be time yet for IORT to replace WBI in early-breast cancer, IORT has been investigated as a strategy for boost in limited-stage breast cancer prior to WBI. Compared to post-operative boost, IORT boost allows precise delivery to a smaller target volume separated from skin, rather than to a volume distended or distorted by seroma, thus improving accuracy and cosmesis. Also, a single-shot boost treatment significantly reduces the duration of adjuvant RT. In a pooled analysis by the International Society of Intraoperative Radiotherapy (ISIORT), IOERT has been demonstrated to be an effective boost strategy with excellent local control rates [42]. A total of 1109 unselected patients belonging to any of the risk groups were treated with IOERT boost (median 10 Gy) followed by WBI (50–54 Gy). At a median follow-up of 6 years, only 16 local recurrences were observed resulting in a local control rate of 99.2%. Grade was the only significant predictor of local recurrence, while none of the age groups demonstrated a higher recurrence rates. Efficacy of KV-IORT as a modality for intraoperative boost has been demonstrated in two large prospective series, an IORT boost of 18–20 Gy was followed by WBI, a local recurrence rate of 1.73% was observed in the study by Vaidya et al [43], while a 3% recurrence rate was seen in the study by Blank et al [44]. The TARGIT-B trial (NCT01792726), which compares EBRT boost versus an IORT boost, in patients at high risk for local recurrence who are receiving breast-conserving treatment, with standard postoperative EBRT has been launched and may provide definite answers.

IORT boost has emerged as an attractive option for boost in combination with oncoplastic surgery [45]. Oncoplastic reconstruction techniques allow for a wider resection margin while maintaining the cosmetic outcome; however, an externally delivered boost, in such cases, has higher chance of partially missing the target volume due to the tissue displacement techniques used for reconstruction. IORT allows for a precise delivery of the radiation boost directly to the tumour bed during surgery and can be followed by oncoplastic reconstruction thus maintaining the oncological safety and improving cosmetic outcome, with other added advantages like avoiding seroma formation and reducing the duration of EBRT. The Breast Centre of the University Hospital of Cologne [45, 46] has recently reported the aesthetic outcomes of X-ray IORT boost (20 Gy) combined with oncoplastic surgery in 149 patients treated since 2011, with excellent cosmetic outcomes in over 90% and seroma formation rates of 2% at 4 weeks.

Colorectal cancers

Locally advanced rectal cancer is best managed with aggressive multimodality treatment involving chemoradiotherapy and radical resection. Most locally advanced (T3) tumours do well with this multimodality approach and local recurrences are seen in only 5–10% of patients. However, in 15% of T4 (unresectable) tumours R0 resections may not be possible [47, 48] and 10% of complete resections still develop local recurrences [47, 48] after full course chemoradiotherapy. Resection status is the most important determinant of local control and survival; incomplete resections yield few long-term survivors. There may be a case for dose escalation in locally advanced/unresectable rectal cancers with incomplete resections or at high risk of local recurrence (close margins); however, gastrointestinal tolerance limits the radiation dose delivered by EBRT. With IORT, higher doses can be delivered directly to the tumour bed without significantly increasing doses to nearby structures. This high dose may be capable of sterilising the margins even after microscopic/macroscopic residual disease.

There is increasing evidence (Table 3) to suggest that inclusion of IORT in the multi-modal treatment of locally advanced rectal cancer can lead to improved local control and survival [11, 4951] especially in the setting of R+ resection. IORT in locally advanced rectal cancer is commonly delivered as an intraoperative boost and used in combination with pre-operative or post-operative radiotherapy with or without chemotherapy [11, 49]. Most studies have utilised IOERT and others have utilised HDR IORT for delivery of IORT in rectal cancer.

Table 3. Studies of IORT in locally advanced colorectal cancer after gross total resection.

Author/Year Sample size Study design T4 % IORT type IORT dose (Gy) EBRT % Median F/U LC 5 year (%) OS 5 year (%) Toxicity grade 3 or >
Ratto et al. [52] 2003 43
IORT-19
No IORT-24
NRC 93 IOERT 10–15 100 74 Sx + IORT: 91
Sx − 57
p = 0.035
61 NR for IORT patients
Sadahiro et al. [53] 2004 IORT-99
No IORT-68
NRC 12 IOERT 15–25 100 (20 Gy only) 67
83
Sx + IORT-98
Sx- 84
p = 0.002
Sx + IORT- 79
Sx- 58
p = 0.002
NR for IORT patients
Ferenschild et al. [54] 2006 123
IORT-30
No IORT-93
NRC 25 HDR-IORT 10 100 25 R0+IORT:72
R0:71 (N.S diff)
R(+) + IORT:58
R (+):0 (p = 0.016)
R0+IORT:56
R0:66 (N.s diff)
R+ ( + ) IORT:38
R+:0 (p= 0.026)
NR for IORT patients
Roeder et al. [55] 2007 243 RC 20 IOERT 10–15 86 59 R0 + IORT- 94
R(+) + IORT-72
NR Total-10%
Proctitis-8
Fistula-7
Bowel stenosis-8
Mathis et al. [50] 2008 146 PC 64 IOERT 7.5–25 100 44 86 52 Total- 22%
Neuropathy-3*
GI/GU-23*
Masaki et al. [56] 2008 44
IORT-19
No IORT-25
RCT 0 IOERT 18–20 No 34 Sx+IORT-94.7
Sx-95.5%
p = 0.344
N.s diff
Urinary catheter indwelling 29% vs. 3%,
Rest- N.s diff
Valentini et al. [57] 2009 100
IORT-29
No IORT-71
NRC 100 IOERT 10–15 100 31 R0+IORT:100
R0:81
p = 0.014
NR NR for IORT patients
Dubois et al [58] 2008 142
IORT-73
No IORT-69
RCT 100 IOERT 15–18 100 60 Sx + IORT- 91.8
Sx- 92.8
p = 0.6018
Sx + IORT - 69.8
Sx- 74.8
p=0.25
No difference in toxicity p=0.15
Kusters et al [59] 2010 605 PC
Pooled analysis
29 IOERT 10–12.5 100 R0+IORT- 90.5
R(+) + IORT-55
p < 0.001
67 NR
Sole et al. [60] 2014 335 PC 16 IOERT 10–15 100 72.6 92 75 Total-10%
GI-19*
GU-8*
Neuropathy-7*
Holman et al [61] 2016 417 PC
Pooled analysis
100%
T4
IOERT 10–12.5 97 52 R0 + IORT- 87
R1 + IORT- 60
R2 + IORT- 57
p < 0.001
R0 + IORT-65
R1 + IORT- 34
R2 + IORT- 14
p < 0.001
NR

NRC: non randomised comparison, RCT: randomised controlled trial, PC: prospective cohort, RC: retrospective cohort, F/U: follow-up, Sx: surgery, R(+): residual after surgery, LC:-local control, OS:-overall Survival, NR: not reported,

*

Number of patients,

N.s diff: Non significant difference.

Table 3 summarises various non-randomised and randomised studies of IORT in locally advanced rectal cancers. The initial non-randomised comparisons [5254, 57, 62] showed conflicting results with IORT after complete resection (R0), while some studies showed equivalent local control [54] in IORT and non IORT group, others showed a significant benefit in local control with IORT [52, 57, 62]. One thing which is certain was that IORT provided significant benefit in local control and survival in patients with R+ resection [54, 55, 59, 61]. The only two randomised studies [56, 58] comparing the addition of IORT to standard treatment failed to show any benefit with addition of IORT in terms of local control or survival. The study by Dubois et al. [58] had a large proportion of T3 tumours (89%), complete resection in most patients would have likely minimised the benefits of IORT, while on the other hand, the study by Masaki et al. [56] was limited by small sample size and inclusion of T1/T2 patients.

Management of locally recurrent rectal (LRRC) cancers presents unique challenges. Prior irradiation in these patients limits the scope for further treatment of these patients with EBRT and is generally associated with poorer survival. IORT with its ability to limit the dose to critical structures serves as a reasonable technique for re-irradiation in LRRC. With the addition of IORT to gross total resection and EBRT, various initial series [63, 64] reported a 5-year survival of over20% even without chemotherapy.

Non-randomised studies of IORT in LRRC (Table 4) have shown a significant improvement in local control with IORT and many series have also shown a survival advantage. Recent series [6570] have also employed Re-EBRT and chemotherapy along with IORT in patients previously treated with pelvic radiotherapy and were able to achieve survival in the range of 30–40%, using these aggressive strategies. Important factors affecting outcomes in most of these studies was completeness of surgical resection [65, 6870] and addition of IORT boost [63, 66, 67]. EBRT during recurrent setting appears to improve the outcomes further and should be considered whenever feasible [68, 70, 71].

Table 4. studies of IORT in locally recurrent colorectal cancers.

Author/Year Sample size Study design IORT type IORT dose (Gy) Prior EBRT % Adj. EBRT % Median Follow up LC 5year (%) OS 5year (%) Toxicity grade 3 or >
Suzuki et al. [63] 1995 106
Sx+IORT:42
Sx:64
NRC IOERT 10–30 25 98 44 Sx+IORT:60%
Sx:7%
at 3years
Sx+IORT:19
Sx: 7%
P=0.0006
Total-36%
Abcess-5
GI/GU-9
wound-3
Valentini et al. [66] 1999 47
Sx+IORT:11
Sx:14
NRC IOERT 10–15 28 100 80 Sx+IORT:80
Sx:24
p < 0.05
Sx+IORT:41
Sx:16
N.S diff
Hydronephrosis-1
Neuropathy-0*
Alektiar et al. [75] 2000 74 RC HDR-IORT 10–18 53 39 22 R0+IORT- 43
R(+) +IORT-26
p = 0.02
R0+IORT-36
R(+) +IORT-11
p = 0.04
Fistula-8*
Neuropathy-1*
Ureter-10*
Wound-5*
Lindel et al. [76] 2001 IORT-49
No IORT-20
NRC IOERT 10–20 14 94 NR R0 + IORT-56
R(+) +IORT-14
R0 + IORT-40
R(+)+IORT-17
Wound complication-4*
Neuropathy-4*
Wiig et al. [67] 2002 107
Sx+IORT:59
Sx:48
NRC IOERT 15–20 0 100 NR Sx+IORT:50
Sx: 30
N.S diff
Sx+IORT:30
Sx: 30%
N.S diff
late toxicity: NR
Acute complication: N.s diff
Dresen et al [69]. 2008 147 RC IOERT 10.–17.5 53 84 NR R0 + IORT-69
R1 + IORT-29
R2 + IORT-28 p < 0.001(3yr)
R0 + IORT-59
R1 + IORT-27
R2 + IORT-24
p < 0.001(3yr)
Neuropathy-16*
Ureter stenosis-4*
Haddock et al. [65] 2011 607 PC IOERT 7.5–30 45 96 44 R0 + IORT-79
R1 + IORT-56
R2 + IORT-49
p < 0.001
R0 + IORT-46
R1 + IORT-27
R2 + IORT-16
p< 0.001
Total-11%
Wound-42*
Neropathy-18*
Roeder et al.[70] 2012 97 PC IOERT 10–20 44 52 33 R0 + IORT-82
R1 + IORT-41
R2 + IORT-18
p < 0.001(3yr)
R0 + IORT-80
R1 + IORT-37
R2 + IORT-35
p < 0.001(3yr)
Acute:
Abscess /fistula-16
Late:
Neuropathy-8*
Ureter stenosis-3*
Calvo et al. [68] 2013 60 RC IOERT 10–15 50 47 36 44
R0 vs R1: HR-2.09, p = 0.05
43
R0 vs R1:HR-2.9, p = 0.05
Total:42%
Fistula-4*
Neuropathy-4*
GI-4*
Holman et al. [71] 2017 565 PC pooled analysis IOERT 10–20 46 95 40 months
In survivors
R0 + IORT-72
R1 + IORT-36
R2 + IORT-39
p < 0.0001
R0 + IORT-48
R1 + IORT-25
R2 + IORT-17
p < 0.0001

NRC: non randomised comparison, RCT: randomised controlled trial, PC: prospective cohort, RC: retrospective cohort, Sx: surgery, R(+): residual after surgery, R0: no residual after surgery, LC: local control, OS: overall survival, NR: not reported,

*

Number of patients,

N.s diff: non-significant difference.

The complication rates in these IORT studies are variable and could range anywhere between 5% and 60%. Wound complications, gastrointestinal problems, ureteric obstruction and neuropathy are some of the frequently encountered morbidities. Wound complications were most common and in some series was quite high, upwards of 40% [50, 62, 72, 73]. Gastrointestinal fistulae and ureteric damage have an incidence ranging from 2% to 12% [50, 62, 64, 72, 73]. Plexopathy and neuropathy are late toxicities of pelvic IORT and have shown a dose-dependent relationship after IORT [50, 62, 64, 65, 72, 73].

A meta-analysis [74] of studies of IORT in locally advanced and recurrent rectal cancers together, has shown a significant benefit with addition of IORT on local control, disease-free survival and overall survival. Meta-analyses of complications did not demonstrate a significant increase in urologic or gastrointestinal complications; however, a greater number of wound complications did occur [74].

Soft-tissue sarcomas

Surgery constitutes the main treatment modality for soft-tissue sarcomas; however, surgery alone cannot provide acceptable local control rates without hampering the functionality of the limb/organ in cases of large and high grade sarcomas, thus making radiation therapy an integral component of function preserving surgery. Radiation therapy used either preoperatively or postoperatively provides acceptable local control rates after an adequate surgery with negative margins. However, in cases of advanced tumours where negative margin is not possible without mutilating surgery (retroperitoneal sarcoma) or in case of recurrent tumours, optimum doses of EBRT cannot be delivered to provide acceptable local control.

IORT has been used in such tumours to escalate doses beyond that of conventional EBRT in an attempt to improve local control rates. In extremity sarcomas, IORT has also been used to replace external boost, reducing the dose and volumes treated with EBRT, so that tolerance of normal structures like joint space, bone, and skin can be respected.

Table 5 summarises studies of IORT in extremity soft-tissue sarcomas, these studies were heterogeneous with varying proportion of recurrent tumours and incomplete resections. Use of IORT in these unfavourable patients aimed at preserving the limb while maintaining acceptable local control. IORT was mostly used in combination with function preserving surgery and moderate doses of EBRT. Recent series [8285] of IORT demonstrate excellent LC rates and functional outcomes, comparable to the series of EBRT alone, despite including higher proportion of tumours with unfavourable factors. Dose of IORT was dependant on resection status, volume and dose of EBRT. While R+ disease fared equally well as R0 disease in the series by Call et al and Kretzler et al [79, 82], studies by Niewald et al and Kretzler et al [79, 81] reported equivalent outcomes in recurrent as well as primary disease. However, in some of the larger series, [8285] resection status and recurrent disease were the most important factors determining local control. Limb preservation was achievable in most patients even with recurrent disease. The complications of neuropathy, contracture, and lymphedema were low, wound complications were the most common complications, and were not much different from that with EBRT [7982].

Table 5. Studies of IORT in extremity soft tissue sarcoma in combination with function preserving surgery and moderate doses of EBRT (40-50Gy).

Author/Year Sample size Study design IORT type IORT dose Gy EBRT % Median follow-up LC 5 year DFS 5 year OS 5 year Complications
Edmonson et al. [77] 2001 39 (Recurrent-3%, R+:38%) Retrospective IOERT/HDR IORT 10–20 100 70 90α NR 80 NR for IORT
Azinovic et al. [78] 2003 45 (Recurrent-42%, R+:33%) Retrospective IOERT 10–20 80 93 80
R0 vs R1:
88 vs 57
p = 0.04
NR 64 Wound complication -4*
Neuropathy-5*
Fracture-2*
Kretzler et al. [79] 2004 28
(Recurrent -57%, R+:39%)
Retrospective HDR/IOERT 12–15 90 55 84 54 66 Total-24%
Neuropathy-1*
Fractures-2*
Contracture-2*
Oertel et al. [80] 2006 153
(Recurrent - 38%, R+: 30%)
Retrospective IOERT 10–20 100 33 78 NR 77 Wound-17%
Neuropathy-7*
Lymphededma-6*
Niewald et al. [81] 2009 38
(Recurrent -24%,)
Retrospective HDR-IORT 8–15 100 27 63 NR 57 Skin-42%
Neuropathy-0
Call et al. [82] 2012 ¥ 61
(Recurrent -21%, R+:18%)
Retrospective IOERT 7.5–20 100 70 91 80 72 Wound-3.2%
Neuropathy-1*
Calvo et al. [83] 2014 159
(R1-16%)
Retrospective pooled IOERT 10–20 100 53 82
R0 vs R1:
p = 0.009
62 72 Acute skin/wound- 16%
Neuropathy-6*
Lympedema-7*
Roeder et al. [84] 2014 34
(R1-12%)
Prospective IOERT 10–15 100 43 97
66 79 Neuropathy-1*
osteonecrosis-1*
Joint dysfunction-1*
Roeder et al. [85] 2016 183
(Recurrent -22%, R1 - 32%)
Retrospective IOERT 8–20 100 64 86
R0 vs R1: 92 vs 75 p = 0.019
61 77 Total-19%
Wound-15*
Neuropathy-14*
osteonecrosis-11*

R+: Residual after surgery, R0: no residual after surgery, LC: local control, OS: overall Survival, DFS: disease-free survival, NR: not reported,

*

Number of patients,

¥

call et al included only upper extremity tumours,

α

-crude rate

Soft-tissue sarcomas in the retro peritoneum are difficult to remove with adequate margins due to their large size, advanced stage, and difficult location with multiple critical organs in close vicinity. Therefore, surgery is often combined with radiotherapy in order to improve the local control rate. However, the proximity of normal organs, such as viscera and neurovascular structures, has made the delivery of therapeutic doses of postoperative EBRT problematic, with higher rates of gastrointestinal complications, including disabling chronic enteritis and fistulae. These difficulties have led to adoption of IORT in the treatment regimen for retroperitoneal sarcoma since the late 1980s.

A randomised trial at the NCI [86], at a median follow-up of 8 years, showed a significantly better local control with IOERT and low-dose post-operative EBRT compared to high-dose post-operative EBRT alone (60% vs. 20%, p < 0.05). The IOERT arm experienced significantly more peripheral neuropathy attributed in part to use of concurrent radio-sensitisers (60% vs. 5%, p < 0.05), while the EBRT only arm had significantly higher GI complications. Experience from other series, summarised in Table 6, has also shown encouraging results with a favourable toxicity profile.

Table 6. Studies of IORT in Retroperitoneal sarcoma.

Author/Year Sample size Study design IORT type IORT dose (Gy) EBRT % Median follow-up LC 5 year (%) OS 5 year (%) Toxicity grade 3 or >
Sindelar et al. [86] 1993 35
GTR-100%
RCT
Sx + IORT + low-dose PORT
Vs Sx + high-dose PORT
IOERT 20 100 96 IORT + PORT- 60%’
PORT-20%
- Neuropathy
IORT + PORT-60%
PORT-5%
Enteritis
IORT + PORT-13%
PORT-50%
Alektiar et al. [87] 2000 32
Recurrent -62%, GTR-94%
Retrospective cohort HDR-IORT 12–15 78 33 62 45 GI -18%
Neuropathy-0%
Gieschen et al. [88] 2001 37
IORT-20
No IORT-17
Recurrent-22%
GTR-78%
Retrospective cohort IOERT 10–20
100
100
38
IORT-83
No IORT-61
p = 0.197¥

IORT-74
No IORT-30
p = 0.044¥
Total-20%
Neuropathy-1*
Fistula-2*
Peterson et al [89] 2002 87
Recurrent-50%, GTR-84%
Retrospective cohort IOERT 8.75–30 89 42 59 47 GI-14%
Neuropathy- 10%
Bobin et al. [90] 2003 24
Recurrent-79%
GTR-92
Retrospective cohort IOERT 8–22 92 53 46 56 Total-8%
Neuropathy-2*
Pierie et al. [91] 2006 IORT-14
No IORT-27
Recurrent-0%
GTR-100%
Retrospective cohort IOERT 10–20 100
100
27 NR IORT-77
No IORT-45
p = 0.38
GI-1%
Neuropathy-3%
Krempien et al. [92] 2006 67
Recurrent-61%, GTR-82%
Retrospective cohort IOERT 12–20 67 30 40 64
R0:87
R+:50
p < 0.01
Fistula-3*
Neuropathy-5*
Urethral stenosis-2*
Pawlik et al. [93] 2006 72
IORT-22
No IORT-50
Recurrent-25%, GTR-75%
Prospective cohort IOERT 15 100 40 60¥ 50 NR
Ballo et al. [94] 2007 83
IORT-18
No IORT-63
Recurrent-28%, R+-47%
Retrospective cohort IOERT 10–15 100 47
IORT-46
No IORT-51
p = 0.9
NR NR
Dziewirski et al. [95] 2010 57
Recurrent -74%, GTR-85%
Prospective cohort HDR-IORT 20 60 20 51 55 NR
Sweeting et al. [96] 2013 18
Recurrent-28%, GTR-100%
Retrospective cohort IOERT 10–20 94 43 64 72 NR
Roeder et al. [97] 2014 27
Recurrent-15%, GTR-100%
Prospective cohort IOERT 10–20 100 33 72 74 Total-6% late toxicity
Stucky et al. [98] 2014 63
IORT-37
Sx only-26
Recurrent-36%,GTR-89%
Retrospective cohort IOERT 10–20
100
0
45
IORT-89
Sx-46
p = 0.03

IORT-60
No IORT-60
Ureteral stricture-1
No grade-3 neuropathy
Gronchi et al. [99] 2014 83
IORT-14pts only
Recurrent-24%
GTR-84%
Prospective cohort IOERT 10–12 88 58 63 59 NR

RCT: randomised control trial, PORT: post-operative RT, Sx: surgery, LC: local control, OS: overall survival, DFS: disease-free survival, NR: not reported,

*

Number of patients

¥

In GTR patients,

α-crude rate

IORT in combination with pre-operative or post-operative RT has shown encouraging results [8794, 97, 99]. While initial reports [87, 89, 90, 92] had higher proportion of patients receiving post-operative RT, recent series [88, 91, 93, 9799] mostly use pre-operative RT because of the smaller volumes that are required with reduced rates of complications. Combination of pre-operative RT, gross total resection and IORT has demonstrated improved local control [91, 98] as well as survival [88, 91] compared to the non-IORT regimens in some of the recent non-randomised comparisons. Resection status and recurrent disease were the most important determinants for local control [89, 92, 94, 97]. GI toxicities, neuropathy and ureteric stenosis are the most common complications with reported rates of 10–35%. They may be dose-dependent, high single dose resulting in greater risk of complications [65, 86, 100].

Most studies of IORT shown in Tables 5 and 6 had a large proportion of recurrent tumours, emphasising the fact that IORT plays a pivotal role in the management of these locally recurrent sarcomas. In a multi-centric, long-term outcomes analysis by the Spanish Cooperative Initiative [101] for Intraoperative electron radiotherapy, 103 patients were investigated to analyse long-term outcomes of locally recurrent soft-tissue sarcoma (LR-STS) patients treated with a multidisciplinary approach. The 5-year IORT in-field control, disease-free survival (DFS), and overall survival were 73%, 43%, and 52%, respectively. Not combining EBRT with surgical resection and IOERT in patients with LR-STS was associated with a significantly increased probability of LR and IOERT in-field relapse. They concluded that low rate of severe toxic events suggests that a multimodality approach with re-resection and IOERT is feasible without prohibitive long-term side effects.

Paediatric tumours

Most paediatric tumours are radiosensitive and radiotherapy constitutes an integral component in their management schema, more so for the unresectable and recurrent tumours, where outcomes remain dismal with chemotherapy alone. However, the use of radiotherapy, especially EBRT in the paediatric population is fraught with late effects like retarded bone and soft tissue growth, abnormal organ development and the risk of second malignancies due to the sensitive nature of these maturing tissues. Thus, there is a narrow therapeutic window within which local control and late effects, which needs to be balanced. The goal of IORT for paediatric tumours is to improve the therapeutic ratio by increasing local control while limiting these late toxicities.

Table 7 summarises various studies of IORT in paediatric tumours, though the numbers are small, IORT has been used across a wide variety of sites and histologies, as a sole radiation modality for radio-sensitive tumours like neuroblastoma [102] or in combination with EBRT for dose escalation to improve local control in sarcomas [103] or for dose de-escalation in RMS with low-dose EBRT. Oertel [103], Goodman [104] and Sole [105] et al included quite a number of recurrent tumours. Use of IORT in combination with surgery and EBRT provided excellent local control across most studies with acceptable toxicity.

Table 7. Studies of IORT in various paediatric tumours.

Author/Year Sample size Study design IORT type IORT dose (Gy) EBRT % Median Follow-up LC 5 year (%) OS 5 year (%) Toxicity grade 3 or >
Haase et al [102] 1994 25 (neuroblastoma) Prospective, single arm IOERT 10–17 NR 51 (mean) 75 63 No late effects at 5-year follow-up
Nag et al [106] 2003 13 (5 metastatic) Retrospective IOERT 10–15 38 42 72
(3yr)
31
(3yr)
late morbidity-30%
Goodman et al [104] 2003 66 (35% recurrent) Retrospective HDR-IORT 12–15 44 12 56
(2yr)
54
(2yr)
Late morbidity-12%
Oertel et al
[103] 2005
18 (17% recurrent) Retrospective IOERT 8–15 100 54.5 95
(3yr)
83
(3yr)
late morbidity-33%
Loss of Limb-1*
Neuropathy-1*
Stauder et al [107] 2011 20 Retrospective IOERT 7.5–25 100 139 77
(10yr)
65
(10yr)
No grade 3 or more late effects or second primary
Sole et al [105] 2015 71 (35% recurrent) Retrospective IOERT 7.5–20 100 72 68
(10yr)
74
(10yr)
Late morbidity-13%
Neuropathy-4*
Necrosis-2*
Lymphedema-2*

LC: local control, OS: overall survival, DFS: disease-free survival, NR: not reported,

*

Number of patients.

In a study by Sole [105] et al, after a median follow-up of 72 months (range, 4–10 months), 10-year LC, disease-free survival, and OS was 74%, 57%, and 68%, respectively. In multivariate analysis after adjustment for other covariates, disease status (p = 0.04 and p = 0.05) and resection margin status (p < 0.01 and p = 0.04) remained significantly associated with LC and OS.

IOERT can be considered as an effective option as a part of multimodality regimen for paediatric solid malignancies, especially for patients with recurrent tumours and abdominopelvic malignancies.

Gynaecological cancers

Recurrent gynaecological malignancies are associated with poor survival due to lack of effective salvage options. Survival rates of locally recurrent cervical cancer after prior radiation therapy are dismal. Most recurrences especially those involving the pelvic sidewall are not resectable and when resection is possible (as in central recurrences), extensive procedures like pelvic exenteration are required, which are associated with a high rate of complications and operative mortality of over 10% [108111]. Introduction of IORT has widened the scope of patients who may be offered surgery and patients who have been previously treated with non-surgical modalities can be offered radical resection when combined with IORT. In resectable recurrences, IORT given after gross total resection can improve local control rates.

IORT has been used to treat locally advanced primary cervical cancers also; however, these series [112] are small and most of the experience comes from recurrent cancers (Table 8). IORT has shown to improve local control and thus survival in locally recurrent cancers [113121] of the uterine cervix and endometrium, limited locoregional recurrences from endometrial cancers doing much better than recurrences from cervical cancers [119, 122124]. The benefit of IORT is seen much more in patients with microscopic residual disease than in those with gross residual disease [113115, 121]. Patient selection based on resection status and volume of recurrence are the most important factors determining outcome after IORT. Previously, irradiated patients when adjusted for resection status and volume of recurrence appear to fare as well as previously un-irradiated patients [115] and addition of EBRT to IORT regimen further improves the control rates [117, 119, 120, 123]. IORT does not seem to increase the rate of acute complications following surgery. Neuropathy and gastrointestinal toxicity are the most common IORT-related toxicities and occur in 5–30% of patients.

Table 8. Studies of IORT in recurrent gynaecological malignancies after gross total resection.

Author/Year Sample size Site Primary/Recurrent IORT type IORT dose (Gy) Prior EBRT % Present EBRT % Median follow-up LC 5yr (%) OS 5yr (%) Toxicity grade 3 or >
Haddock et al. [113, 115] 1996 63 Cervix-40
Endometrium-16
Others-8
Primary-16%
Recurrent-84%
IOERT 8–25 0
70
100
63
NR 61 27 Total-17%
GI-5*
Neuropathy-2*
Mahe et al. [124, 125] 1996 70 Cervix Recurrent-100% IOERT
X ray IORT
10–30 97 42 15 21 (3yr) 8 (3yr) Total-14%Neuropathy-5*
Ureteral obstruction-4*
Del carmen et al. [126] 2000 15 Cervix-5Endometrium-3
Vagina-7
Recurrent-93% IOERT 10–22.5 60 6 NR NR 74 Neuropathy-4*
GU-3*
Lymphedema-2*
Martinez-monge et al.[116] 2001 67 Cervix Primary-46%
Recurrent-54%
IOERT 10–25
10–20

86
97
14
58
19
80.5
49
58 (10yr)
14 (10yr)
Total-14.9%
Neuropathy-1*
Chronic pain-8*
Gemignani et al. [114] 2001 17 Cervix-9
Endometrium-7
Vagina-1
Recurrent-
100%
HDR-IORT 12–15 82 12 20 67 (3yr) 54 (3yr) GI-4*
Neuropathy-3*
Wound-4*
Dowdy et al. [126] 2006 25 Endometrium Recurrent-
100%
IOERT 10–25 56 84 34 84 R0-71
R1-47
R2-0
Neuropathy-8*
GU-5*
Fitula-5*
Tran et al. [118]
2007
36 Cervix-17
Endometrium-11
Others-8
Recurrent-89% X-ray IORT 6–17.5 72 53 50 44 47 Total-27%
Wound-4*
GI-1*
Barney et al [119].2013 86 Cervix-100 Recurrent-85% IOERT 6–25 81 71 32 Primary-70
Recurrent-61%
(3yr)
25
(3yr)
GI-4*
GU-4*
Neuropathy-1*
Calvo et al.[120] 2013 35 Cervix-20 Endometrium-7 Others-8 Recurrent-
100%
IOERT 10–15 71 46 46 58 42 Fistula-5*
Ureter stenosi-2*
Neuropathy-1*
Backes et al.[127] 2014 32 Cervix-21
Others-11
IORT in 66% only
Recurrent-
100%
IOERT/HDR-IORT 10–20 100 0 NR PE+IORT-10Mon
LEER+IORT-9Mon
PE-33Mon
PE+IORT-10Mon
LEER+IORT-10Mon
PE-41Mon
NR
Foley et al.[121] 2014 32 Cervix-21
Endometrium-6
Others-5
Rccurent-81% IOERT 10–22.5 88 20 26 R1-73
R2-71
R1-77
R2-55 p = 0.001
Total-47%
GU-2
Lymphedema-2
Sole et al. [117] 2014 61 Cervix-18
Endometrium-32
Ovarian-9
Vagina-2
Recurrent
Pelvic-57%
Para-aortic-
43%
IOERT 10–15 66 48 42 65 45 Total-20%
Fistula-5*
Neuropathy-1*
Wound-3*
Arians et al.[122] 2016 36 Cervix-18
Endometrium-12
Vulva-6
Recurrent-
100%
IOERT 10–18 76
25
100
11
17
17
14 Cervix-0
Endometrium-40
Vulva-20
p = 0.017
Cervix-6
Endometrium-50
Vulva-17
P = 0.038
Wound complications-5*
Neuropathy-4*
Lymphedema-3*

Sx: surgery, LC: local control, OS: overall survival, NR: not reported,

*

Number of patients

PE: pelvic exenteration, LEER: laterally extended endopelvic resection, Mon: median survival in months.

Genitourinary cancers

Bladder cancer

Although multiple reports of perioperative brachytherapy in bladder cancer are available with encouraging results, there is limited data on IORT in bladder cancer, with only one small retrospective series in recurrent bladder cancer meeting our search and selection criteria. Recurrent bladder tumours after a cystectomy are associated with dismal survival rates, owing to the fact that adequate surgery is often not feasible and salvage with high doses of EBRT is difficult due to the tolerance of adjacent organs. IORT is used to deliver high doses to the tumour in an effort to improve local control. Hallemeier et al [128] reported the use of IOERT in 17 patients after maximal resection of disease. Pre- or post-operative EBRT was used in 94% of patients. Encouraging 2-year local control and survival was seen, completely resected tumours were associated with a significant improvement in survival compared to gross residual disease.

Renal cancer

Radical surgery forms the mainstay of treatment in patients with renal cell carcinoma (RCC). However, in patients with recurrent and advanced tumours, achieving complete resection with wide margins may be difficult due to proximity to the critical structures and this effects not only the local control but survival as well [129]. Adjuvant EBRT in this setting may improve local control; however, the doses achievable with EBRT is limited due to low tolerance of the surrounding structures like stomach, small bowel, contralateral kidney, liver, and spinal cord. IORT offers an attractive treatment option to escalate doses to the tumour bed, especially in cases with positive resection margins. Studies evaluating the role of IORT in the management of locally advanced and recurrent RCC are summarised in Table 9 [129134].

Table 9. Studies of IORT in bladder and renal cancers.
Author/Year Sample size Study design IORT type IORT dose (Gy) Prior EBRT % Present EBRT % Median follow-up months LC 5 year % OS 5 year % Toxicity grade 3 or >
Hallemeier et al. [128] 2013 Bladder-13, Ureter-4
Recurrent-88%
Retrospective cohort IOERT 10–20 24 94 43.2
In survivors
51
(2 yr)
16 (5 yr)
R0+R1: 56
R2-11
p = 0.03 (2 yr)
Total-12%
Ureter stricture-4
Fistula-1
Paly et al. [134] 2014 98-RCC
Recurrent-72%
Retrospective IOERT 9.5–20 63 42
In survivors
76 Advanced-37
Recurrent-55
Total-5%
Pancreatic leak-3*
gastritis-1*
ARDS-1*
Calvo et al. [130] 2013 25-RCC
Recurrent-40%
Retrospective IOERT 9–15 - 60 266 80 38 Total-24%
Habl et al. [132] 2013 17-RCC
Recurrent-100%
Retrospective IOERT 10–20 65 18 91 (2 yr) 73
(2 yr)
None
Hallemeier et al. [129] 2012 22-RCC
Recurrent-86%
Retrospective IOERT 10–20 - 95 119
In survivors
73 40
R0-80
R1-29
p = 0.057
Total-23%
ARDS-1* Pancreatic pseudocyst-1*
Perforated ulcer-2*
Master et al. [133] 2005 14-RCC
Recurrent-100%
Retrospective IOERT 12–20 NR 66
(mean in survivors)
85
(crude rate)
30 NR
Eble et al. [131] 1998 11-RCC
Recurrent-73%
Retrospective IOERT 15–20 - 100 24 100 47
(4 yr)
Wound-2*
None IORT related

RCT: randomised control trial, Sx: surgery, LC: local control, OS: overall survival, DFS: disease-free survival, NR:-not reported,

*

Number of patients

ARDS: acute respiratory distress syndrome.

Habl et al [132] reported outcomes with IOERT after complete surgical resection in a cohort of 17 patients with locally recurrent RCC. Although R0 resection could be achieved in only one-third of the patients, most patients failed distally, with only two local recurrences. None of the patients suffered from any acute or late radiation toxicities. One of the largest series of IOERT in RCC has been reported by Paly et al in a multi-institutional cohort of 98 patients. Twenty-eight per cent patients had advanced disease at presentation and 72% had recurrent disease. More than 50% had residual disease after resection. Sixty-two per cent received additional pre-operative or post-operative EBRT. An excellent local control of 72% at 5 years was demonstrated with grade 3 toxicity in 5% of patients. Higher IORT dose was associated with improved survival (p < 0.001). Thus, studies of IORT in RCC, though retrospective in nature demonstrate a consistently high local control rate in recurrent/advanced RCC with acceptable toxicity rates.

Prostate cancer

Locally advanced/high-risk prostate cancer is associated with significant risk of relapse when treated with radical prostatectomy alone, risk being the highest when the margins are positive. Adjuvant radiotherapy in this setting reduces the risk of relapse significantly [135]. IORT has been explored in high-risk prostate cancers in combination with radical prostatectomy and post-operative EBRT to improve local control via dose escalation. IORT has the added radiobiological advantage of high single dose of radiation, which improves the therapeutic gain due to low α/β of prostate. It also helps limit doses to the rectum and has been shown to have low gastrointestinal (GI) morbidity even in combination with EBRT [136]. Several small prospective series (Table 10 [136141]) have evaluated the feasibility of this multi-modality approach in patients with non-metastatic, node-negative disease with probability of LN involvement being less than 15%. Encouraging local control and acceptable toxicity has been demonstrated even though significant proportion of patients had margin positive disease in these series [136, 139, 141]; however, long-term results are awaited.

Table 10. IORT studies for prostate cancers.
Author/Year Sample size Study design IORT type IORT dose (Gy) Present EBRT % Median follow-up months LC 5 year % OS 5 year % Toxicity grade 3 or >
Krengli et al. [139] 2010 38 (intermediate–
high-risk Pca,
Margin +ve: 71%)
Prospective IOERT 10–12 Margin +ve and/or ECE 18.2 NR NR
No grade 3
complications
Rocco et al. [136] 2009 IORT-33
(intermediate–
high-risk Pca,
Margin +ve: 24%)
RP-100
Matched pair analysis IOERT 12 IORT-88
R1, pT4, N+
RP-44
16 IORT-97
RP-86 (bRFS)
100
( 2yr)
Acute grade ≥ 2
IORT vs. RP
GU-7% vs. 5%
GI-3% vs. 4%
Lategrade ≥ 2
IORT vs. RP
GU-3% vs. 1%
GI-0% vs. 1%
Saracino et al [141] 2007 34
(intermediate-risk Pca, Margin +ve: 41%)
Prospective IOERT 16–22 None 41 77.3
(3-yr bRFS)
71
(3-yr)
None
Orrechia et al. [140] 2007 11
(high-risk Pca)
Prospective IOERT 12 67 NR NR NR 1 had acute symptomatic lymphocele
Kato et al. [138] 1998 54
Stage B2-D1α
No RP
Prospective IOERT 25–30 100
(30 Gy)
54 83 (LC)
75
(bRFS)
NR Late rectal toxicity-7%
(No toxicity with IORT of 25 Gy)
Higashi et al. [137] 1998 35
Stage B-Cα
No RP
Prospective IOERT 25–30 100
(30 Gy)
NR NR Stage B-92
Stage C-87
NR

RCT: randomised control trial, Sx: surgery, LC: local control, OS: overall survival, DFS: disease-free survival, NR: not reported, *Number of patients, RP: radical prostatectomy, Pca: prostate cancer, ECE: extra capsular extension, R1: margin +ve, N+: node-positive disease, bRFS: biochemical relapse-free survival,

α

Whitmore-Jewett staging system [Whitmore 1956, Jewett 1975].

Upper gastro-intestinal tumours

Gastric cancers

Curative resection is the mainstay of treatment for gastric cancer; however, high incidence of locoregional and systemic failures, makes outcomes dismal, especially in cases with gastric serosal involvement and/or nodal involvement [142, 143]. Attempts to improve locoregional control and survival include addition of adjuvant radiotherapy/chemoradiation [144], perioperative chemotherapy [145] and extensive surgeries including D2/D3 resections [146, 147]. Despite significant improvements in disease control and survival with adjuvant chemoradiotherapy, local and regional recurrences remain high at 19% and 65%, respectively, after tri-modality therapy [144]. Therefore, there may be a case for dose escalation with IORT in advanced gastric carcinomas (especially serosal/nodal involvement) to improve local/regional control. IORT in gastric cancer involves boosting the tumour bed, remaining lymphatic networks, and nodal basins to control residual microscopic disease and improve locoregional control.

Role of IORT in gastric cancer after curative resection has been evaluated in multiple studies (retrospective, prospective, and randomised control), which have shown an improvement in locoregional control [148, 150] and survival with IORT, especially in patients with stage-II/stage-III and node-positive disease (Table 11) [1, 148, 150154]. While initial studies of IORT involved less aggressive surgeries (D1) and infrequent use of adjuvant radiotherapy recent studies [148, 150, 153] have demonstrated a consistent benefit with IORT, even in combination with D2 resections and post-operative CTRT. Extended resections like D3 may reduce the benefit with IORT [153], however, IORT combined with a limited lymph node dissection (D1) may be associated with survival similar to extended dissection (D2/3), with lesser post-operative mortality [156]. While most studies did not show an increase in complications with the use of IORT, Drognitz et al [151] have demonstrated a significant increase in surgical complications with the use of IORT (44% vs. 20%, p < 0.05). They also did not show a benefit with addition of IORT to surgical resection. Complication rates need to be carefully weighed against improvement in locoregional control to maximise benefits with IORT [157].

Table 11. IORT studies for gastric cancers.
Author/Year Sample size Study design IORT type IORT dose (Gy) EBRT (%) Type of nodal dissection Median follow-up (months) LRC OS 5 year (%) Toxicity grade 3 or >
Sindelar et al. [149] 1993 Sx+IORT-27
Sx+/-EBRT-33
RCT IOERT 20 0
72
NR 84 37
8
p < 0.001
25 months (M.S)
21 months (M.S)
p = 0.99
Fistula:
IORT-4 vs. Sx-5*
Enteritis:
IORT-0 vs. Sx-2*
Abe et al [1] 1995 IORT-94
No IORT-127
NRC IOERT 28–35 none NR NR NR Stage II:
IORT-78
No IORT-66
Stage III:
IORT-60
No IORT-51
Stage IV:
IORT-33
No IORT-14 (all N.S)
NR
Avizonis et al.[158] 1995 27 Prospective phase II IOERT 12.5–16.5 79 NR NR 85 47
(2yr)
Acute
toxicity-14%
Late-7%
Ogata et al. [152] 1995 IORT-58
No IORT-120
Retrospective IOERT 12 None D2 NR NR Stage II:
IORT-100,
No IORT-63 (4yr)
Stage III:
IORT-55
No IORT-35 (8yr)
Stage IV:
IORT-12
No IORT-13 (5yr)
None
Coquard et al. [156] 1997 63
(R0-92%)
Retrospective IOERT 12–23 48 D1-89% 61 in survivors 76 (crude rate) 47 None attributed to IORT
Skoropad et al. [154] 2000 78
Pre-op RT+IORT+ Sx vs. Sx alone
RCT IOERT 20 100
(Pre-op RT-20 Gy/5#)
D1 NR NR Entire cohort:
IORT:21months
No IORT: 9months
(P = 0.311)
Node +ve and
Advanced stage: IORT vs. No IORT:
p < 0.05
Similar acute toxicity in both arms
Higher pancreatitis surgery alone.
No RT late toxicity
Weese et al. [159] 2000 16
(IORT-56%)
Prospective IOERT 10 88 D2 27 93 (crude rate) 66 (crude rate at 3yr) NR
Glehen et al. [160] 2003 42
(All Node +ve, R0-93%)
Retrospective IOERT 12–15 97 NR 131 79 45 NR
Miller et al. [161] 2006 50
(R0-42%, recurrent-26%, oesophagus-14%)
Retrospective IOERT 10–25 96 NR 19 75 15 (5 yr) Acute-48%
Chronic-26%
GI-6*
Overall treatment-related mortality-6%
Qin et al. [153] 2006 IORT-106
No IORT-441
NRC IOERT 10–30 None D2/3 NR NR Stage III D2:
IORT- 60%
No IORT-36%
p < 0.005
Stage III D3:
IORT-61%
No IORT-56%
p > 0.05
NR
Drognitz et al. [151] 2007 IORT-61
No IORT-61
(R0-100%)
NRC IOERT 15–25 None D2 56 90 IORT-58%
No IORT-59%
p = 0.99
Perioperative mortality-4.9% both groups
Surgical morbidity:
IORT:44%
No IORT: 20%
Fu et al. [148] 2008 97
IORT-46
No IORT-51
(R0-90%)
Prospective IOERT 12–15 100%
CTRT
D2 24 77
IORT- 77
No IORT- 63
p = 0.05, (3Yr)
44
IORT-56
No IORT-47
p = 0.20, (3Yr)
Late toxicity-3*
No difference among groups
Zhang et al. [150] 2012 97
IORT-46
No IORT-51
(R0-90%)
Prospective IOERT 12–15 100% CTRT D2 37 50
IORT-50
No IORT-35
p = 0.04
26
IORT-28
No IORT-26
p = 0.4
IORT vs. No IORT
Acute-39% vs. 37%
Late-.10% vs. 0%, p = 0.02
Enteritis-1*
Haemorrhage-4*
Calvo et al. [155] 2012 32
(R0-100%)
Retrospective IOERT 10–15 47 D2 40 84 55 Acute GI-5*

RCT: randomised control trial, NRC: non-randomised comparison, Sx: surgery, LC: local control, OS: overall survival, DFS: disease-free survival, NR: not reported,

*

Number of patients,

#

fractions,

Pre-op: pre-operative, M.S: median survival, N.S: non-significant.

Pancreatic cancer

Pancreatic cancer is associated with dismal survival rates even in completely resected patients. Significant proportion of patients either develop locoregional recurrence or systemic metastases. Multi-modality treatment approaches combining chemotherapy and radiotherapy in addition to surgery have resulted in some improvement in locoregional control and survival [162164]. Attempts at radiotherapy dose escalation with EBRT have been limited due to the location of tumour. IORT can result in delivery of higher doses to the tumour bed and may improve local control and survival in resected pancreatic cancers. In unresectable tumours, IORT alone or in combination with EBRT can provide some local control along with effective palliation of symptoms.

Studies of IORT in resectable pancreatic cancers are summarised in Table 12 [165171], though heterogeneous in proportion of R1 resections and use of adjuvant EBRT and/or chemotherapy, they have been consistent in showing an improvement in locoregional control [165168]. Some studies have also shown an improvement in survival [165, 168, 172, 173]. Addition of IORT to standard treatment did not result in any increase in perioperative morbidity or late toxicity rates [165, 167169, 173]. Stage [172, 173], R0 resection [166], chemotherapy [170], and pre-operative treatment [168] were other important determinants of survival in these studies. A systematic review also agreed with observations from these non-randomised studies and suggested a survival benefit with IORT in resected pancreatic patients.

Table 12. Studies of IORT in the management of pancreatic cancers.
Author/Year Sample size Study design IORT type IORT dose (Gy) EBRT % Median follow-up LC 5 year (%) OS 5 year (%) Toxicity grade 3 or >
Mohiuddin et al. [178] 1995 49 UR-PC
(Resected-0%)
Retrospective IOERT 10–20 100
(Post-op)
28 69 7 (4yr) Acute toxicity-14%
GI bleeding-2*
Late toxicity-19%
GI bleeding-3*
Obstruction-2*
Nishimura et al. [179] 1997 Resected-157
IORT- 55
No IORT-102
Unresectable-175
IORT-71
No IORT-104
Retrospective IOERT 12–33 Gy Resected-
70
Unresectable-87
(Pre-op or post-op)
NR NR Resected:
IORT-16
No IORT-0 (2yr)
Unresectable:
IORT-14
No IORT-0
p < 0.05 (2yr)
IORT:
Late toxicity:
gastric ulcer-18*
Intestinal perforation-4*
Ma et al. [177] 2004 81 UR-PC
(Resected-0%)
IORT-18
IORT+EBRT-25
EBRT-16
Palliative Sx-22
Retrospective IOERT 15–25 80
(Post-op)
NR NR
(60% complete pain relief with IORT)

10.7 (M.S)
12.2 (M.S)
5.1 (M.S)
7 (M.S)
IORT vs IORT+EBRT:
Delayed gastric
emptying: 3 vs 2*
Willet et al. [180] 2005 150 UR-PC
(Resected-0%)
Retrospective IOERT 15–20 Gy 100 (Pre/post-op RT) NR NR 7 (3Yr) Post-operative complications- 20%
Late toxicity- 15%
Upper GI bleed-16*
Jingu et al. [176] 2012 322
Resected-83
Unresectable- 109
Metastatic-130 (exclude)
Retrospective IOERT 20–30 29 (post-op RT) 38 64 9
R2:HR-2.03,
p < 0.001
Late toxicity
GI-4*
Cai et al. [174] 2013 194 UR-PC
(Resected-0%)
Retrospective IOERT 10–25 97% (Pre-op CTRT) 12 38 (3yr) 6 (3yr) Acute toxicity- 21%
Late toxicity-14%
Haemorrhage-23*
Chen et al. [175]
2016
247 UR-PC
(Resected-0%)
Retrospective IOERT 10–20 51% (Post-op CTRT) 10 35 (3yr)
Complete pain relief-70%
7.2 (3yr) Post-operative complications-14%
Fistula-11*
Haemorrhage- 7*
Keane et al. [181] 2016
68 UR-PC
After NACTRT
Resected-41 IORT-22 (R1-73%)
No IORT-19
Unresectable-18 (IORT-17)
Retrospective IOERT
8–13
15–17
100 % (NACTRT) 21 NR
35.1 (M.S)
24.5 (M.S)
24.8 (M.S)
No significant difference in post-operative complications
Kokubo et al. [172] 2000 138 R/BR-PC
(Resected-100%, R1-29%)
Retrospective IOERT 20–30 45
(Pre-op:13%
Post op-47%
Both-40%)
NR NR R0:19
R1:4
p < 0.005
(2-yr cause specific survival)
Acute toxicity- none
Late toxicity-
GI ulcers-20%
Perforation-2*
Alfieri et al. [165] 2001 46 R/BR-PC (Resected-100%)
IORT-26 (R1-10%)
No IORT-20 (R1-13%)
NRC IOERT 10
100
0
(Post-op)
82
IORT-58
No IORT-30
p < 0.001

IORT-16
No IORT-6
p = 0.06
IORT vs. No IORT
Acute morbidity-
57% vs. 43% (p = 0.1)
Perioperative mortality-
8% vs. 9%
Reni et al. [173] 2001 127 R/BR-PC
(Resected-82%)
IORT- 127
(R0-1:104, R2-23)
No IORT- 76 (R0-1:62, R2-14)
Retrospective IOERT 10–25
32
20
(Post-op)
21 (in
survivors)
Stage I-II:
IORT-73
No IORT-40
Stage III-IVA:
IORT-50
No IORT-45
Stage I–II:
IORT-22
No IORT-6
Stage III-IVA:
IORT-3
No IORT-5
IORT vs. No IORT: Acute toxicity: N.S difference
Chronic toxicity:
Abdominal pain 15 vs. 22%
Late GI bleed-6 vs. 3%
Stenosis-3% vs. 0%
Messick et al. [169] 2008 49 R/BR-PC (Resected-100%, R1-74%)
IORT-22
No IORT-27
NRC IOERT 10–12
76
64

10.1
13.3

IORT-82
No IORT-88
(N.S)

IORT-20 (M.S)
No IORT-13 (M.S)
(N.S)
IORT vs. No IORT: Delayed gastric emptying-6.7 vs. 4.2%
Wound infection-4.5 vs. 22%
Pancreatic fistula 10 vs. 4.8% (N.S)
Valentini et al. [171] 2008 26 R/BR-PC
Resected-100%, R1-4%
Retrospective IOERT 10 100
(Post-op: 65%)
102 in survivors 57 15 Perioperative
complications-11%
Showalter et al. [167] 2009 R/BR-PC
IORT-37 (R1-2:43%)
No IORT-46 (R1-2: 30%)
Retrospective IOERT 10–20 74
66
(Post-op RT)
NR IORT-79
No IORT-61
p = 0.19
IORT-21 (M.S)
No IORT-19 (M.S)
Perioperative
complications-46% vs. 40% (N.S)
Valentini et al. [168] 2009 270
Resected-81%
(R1-27%)
ISIORT Pooled analysis IOERT 7.5–25 64
(pre-op:24
Post-op:40)
96 23
Pre-op RT vs. post-op RT vs. IORT alone
p < 0.0001
18
IORT + Pre-op RT vs. IORT + post-op RT vs. IORT alone
p < 0.0001
Acute toxicity- None > G2
Late-NS
Ogawa et al. [170] 2010 210 R/BR-PC
Resected-100%, R1-32%
Retrospective IOERT 20–30 30 26 84 (2yr) 42 (2yr) Late toxicity
GI-7*
Calvo et al. [166]
2013
60 R/BR-PC
(Resected-83%,
R1-43%)
IORT-29
No IORT-31
Prospective IOERT 10–15 100%
(Pre-op CTRT-32%)
16 58
No IORT:
HR-6.75,
p = 0.01
20
Perioperative complications-43% (N.S)
Chronic-17%
Neuropathy-4*
GI-3*

RCT: randomised control trial, NRC: non-randomised comparison, Sx: surgery, LC: local control, OS: overall survival, DFS: disease-free survival, NR: not reported,

*

Number of patients,

# fractions, Pre-op: pre-operative, Post-op: post-operative, M.S: median survival in months, N.S: non-significant, R/BR-PC: resectable/borderline resectable pancreatic cancer, UR-PC: unresectable pancreatic cancer, NACTRT: neoadjuvant chemoradiotherapy, Resected: complete resections (R0/R1), Unresectable: R2/palliative resections.

Studies of IORT in unresectable pancreatic cancer (Table 12 [174180]) on the other hand, have failed to demonstrate a survival benefit with the addition of IORT, though an improved local control was seen [174176, 178, 181]. IORT also resulted in significant pain relief and palliation of symptoms [175, 177179] with no additional morbidity or toxicity [175, 177, 181]. Tumour size [17, 174, 176, 180], metastasis [179], and chemotherapy [17, 174, 176] were predictors of survival in these studies of unresectable pancreatic cancer. Most of these studies included patients treated before the year 2000 and utilised post-operative radiotherapy and chemotherapy with older regimes. In the current era, pre-operative chemotherapy (± radiotherapy) with novel systemic agents (like FOLFIRINOX and nab-Paclitaxel) has shown to improve resectablity rates and survival in unresectable pancreatic cancers [182, 183]. Keane et al [181], evaluated the role of IORT in combination with intensive neoadjuvant chemoradiotherapy regimens and demonstrated encouraging survival rates in patients with close/positive margins and unresectable disease with no increase in toxicity. Further studies are required to better define the role of IORT in the management of pancreatic cancers, in the current era especially with the advent of novel systemic agents.

Conclusion

Intraoperative radiation therapy is an attractive treatment option for patients with colorectal, gynaecological, intra-abdominal, head and neck, and most recently, breast cancers. IORT has been used in a multitude of roles across these sites, for dose escalation, EBRT dose de-escalation, as sole radiation modality in early-breast cancers and as a Re-irradiation modality in recurrent cancers. IORT serves its role best in combination with gross total resection and moderate doses of EBRT. Utility of IORT has been tested in the setting of a randomised control trial in early breast, retroperitoneum, gastric and colorectal cancers, the results of which support the use of IORT as a management option in these settings. However, appropriate technique and patient selection is the key to success with IORT. IORT has the potential to improve outcomes in recurrent cancers of the pelvis, head and neck and colorectum and can be considered as a supplement to gross total resection. In paediatric tumours, IORT serves to decrease late toxicities associated with EBRT. In appropriately selected patients, complication rates associated with IORT are low.

References

  • 1.Abe M, Shibamoto Y, Ono K, et al. Intraoperative radiation therapy for carcinoma of the stomach and pancreas. Front Radiat Ther Oncol. 1991;25:258–69. doi: 10.1159/000429597. [DOI] [PubMed] [Google Scholar]
  • 2.Abe M, Takahashi M. Intraoperative radiotherapy: the Japanese experience. Int J Radiat Oncol Biol Phys. 1981;7(7):863–8. doi: 10.1016/0360-3016(81)90001-8. [DOI] [PubMed] [Google Scholar]
  • 3.Abe MFM, Yaniano K, et al. Intraoperative irradiation in abdominal and cerebral tumours. Acta Radiol. 1971;10:408–16. doi: 10.3109/02841867109130786. [DOI] [PubMed] [Google Scholar]
  • 4.Intraoperative radiation therapy. Proceedings of the third international symposium on intraoperative radiation therapy. In: Abe M, Takahashi M, editors. 1991. [Google Scholar]
  • 5.Goldson A. Past, present and prospects of intraoperative radiotherapy (IOR) Semin Oncol. 1981 [PubMed] [Google Scholar]
  • 6.Veronesi U OR, Luini A, et al. A preliminary report of intraoperative radiotherapy (IORT) in limited-stage breast cancers that are conservatively treated. Eur J Cancer. 2001;2001(37):2178–83. doi: 10.1016/S0959-8049(01)00285-4. [DOI] [PubMed] [Google Scholar]
  • 7.Vaeth JMea. Intraoperative radiation therapy in the treatment of cancer. Front Radiat Ther Oncol. 1996;31:65–7. [Google Scholar]
  • 8.Harrison LB EW, Anderson LL. High dose rate intraoperative radiation therapy for colorectal cancer. I Oncol. 1995;9:679–83. [PubMed] [Google Scholar]
  • 9.Harrison LB EW, Anderson LL. High-dose rate intraoperative radiation therapy for colorectal cancer: II. Oncol. 1995;9:737–41. [PubMed] [Google Scholar]
  • 10.Bratengeier K KT. Homogeneous Ir-192 afterloading-flab irradiation of plane surfaces. Z Med Phys. 2002;12(230–7) doi: 10.1016/S0939-3889(15)70477-0. [DOI] [PubMed] [Google Scholar]
  • 11.Huber FT SR, Zimmerman F, et al. Locally advanced rectal cancer: resection and intraoperative radiotherapy using the flab method combined with preoperative or postoperative radiochemotherapy. Dis Colon Rectum. 1996;39(774–9) doi: 10.1007/BF02054443. [DOI] [PubMed] [Google Scholar]
  • 12.Bernier J, Cooper JS, Pajak TF, et al. Defining risk levels in locally advanced head and neck cancers: a comparative analysis of concurrent postoperative radiation plus chemotherapy trials of the EORTC (#22931) and RTOG (# 9501) Head Neck. 2005;27(10):843–50. doi: 10.1002/hed.20279. [DOI] [PubMed] [Google Scholar]
  • 13.Bernier J, Domenge C, Ozsahin M, et al. Postoperative irradiation with or without concomitant chemotherapy for locally advanced head and neck cancer. N Engl J Med. 2004;350(19):1945–52. doi: 10.1056/NEJMoa032641. [DOI] [PubMed] [Google Scholar]
  • 14.Cooper JS, Zhang Q, Pajak TF, et al. Long-term follow-up of the RTOG 9501/intergroup phase III trial: postoperative concurrent radiation therapy and chemotherapy in high-risk squamous cell carcinoma of the head and neck. Int J Radiat Oncol Biol Phys. 2012;84(5):1198–205. doi: 10.1016/j.ijrobp.2012.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Pignon JP, Bourhis J, Domenge C, et al. Chemotherapy added to locoregional treatment for head and neck squamous-cell carcinoma: three meta-analyses of updated individual data MACH-NC collaborative group Meta-analysis of chemotherapy on head and neck cancer. Lancet. 2000;355(9208):949–55. doi: 10.1016/S0140-6736(00)90011-4. [DOI] [PubMed] [Google Scholar]
  • 16.Janot F, de Raucourt D, Benhamou E, et al. Randomized trial of postoperative reirradiation combined with chemotherapy after salvage surgery compared with salvage surgery alone in head and neck carcinoma. J Clin Oncol. 2008;26(34):5518–23. doi: 10.1200/JCO.2007.15.0102. [DOI] [PubMed] [Google Scholar]
  • 17.Chen AM, Bucci MK, Singer MI, et al. Intraoperative radiation therapy for recurrent head-and-neck cancer: the UCSF experience. Int J Radiat Oncol Biol Phys. 2007;67(1):122–9. doi: 10.1016/j.ijrobp.2006.08.038. [DOI] [PubMed] [Google Scholar]
  • 18.Nag S, Schuller DE, Martinez-Monge R, et al. Intraoperative electron beam radiotherapy for previously irradiated advanced head and neck malignancies. Int J Radiat Oncol Biol Phys. 1998;42(5):1085–9. doi: 10.1016/S0360-3016(98)00289-2. [DOI] [PubMed] [Google Scholar]
  • 19.Perry DJ, Chan K, Wolden S, et al. High-dose-rate intraoperative radiation therapy for recurrent head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2010;76(4):1140–6. doi: 10.1016/j.ijrobp.2009.03.025. [DOI] [PubMed] [Google Scholar]
  • 20.Scala LM, Hu K, Urken ML, et al. Intraoperative high-dose-rate radiotherapy in the management of locoregionally recurrent head and neck cancer. Head Neck. 2013;35(4):485–92. doi: 10.1002/hed.23007. [DOI] [PubMed] [Google Scholar]
  • 21.Zeidan YH, Shiue K, Weed D, et al. Intraoperative radiotherapy for parotid cancer: a single-institution experience. Int J Radiat Oncol Biol Phys. 2012;82(5):1831–6. doi: 10.1016/j.ijrobp.2011.02.033. [DOI] [PubMed] [Google Scholar]
  • 22.Zeidan YH, Yeh A, Weed D, et al. Intraoperative radiation therapy for advanced cervical metastasis: a single institution experience. Radiat Oncol. 2011;6:72. doi: 10.1186/1748-717X-6-72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Pinheiro AD, Foote RL, McCaffrey TV, et al. Intraoperative radiotherapy for head and neck and skull base cancer. Head Neck. 2003;25(3):217–25. doi: 10.1002/hed.10203. [DOI] [PubMed] [Google Scholar]
  • 24.Vaidya JS BM, Tobias JS, et al. Targeted intra-operative radiotherapy (Targit): an innovative method of treatment for early breast cancer. Ann Oncol. 2001;12 doi: 10.1023/A:1011609401132. [DOI] [PubMed] [Google Scholar]
  • 25.Vaidya JS BM, Tobias JS, et al. Targeted intraoperative radiotherapy (TARGIT) yields very low recurrence rates when given as a boost. Int J Radiat Oncol Biol Phys. 2006;66:1335–38. doi: 10.1016/j.ijrobp.2006.07.1378. [DOI] [PubMed] [Google Scholar]
  • 26.Veronesi U OR, Luini A, et al. Intraoperative radiotherapy during breast conserving surgery: a study on 1,822 cases treated with electrons. Breast Cancer Res Treat. 2010;124:141–51. doi: 10.1007/s10549-010-1115-5. [DOI] [PubMed] [Google Scholar]
  • 27.Vaidya JS, Wenz F, Bulsara M, et al. Risk-adapted targeted intraoperative radiotherapy versus whole-breast radiotherapy for breast cancer: 5-year results for local control and overall survival from the TARGIT-A randomised trial. Lancet. 2014;383(9917):603–13. doi: 10.1016/S0140-6736(13)61950-9. [DOI] [PubMed] [Google Scholar]
  • 28.Veronesi U, Orecchia R, Maisonneuve P, et al. Intraoperative radiotherapy versus external radiotherapy for early breast cancer (ELIOT): a randomised controlled equivalence trial. Lancet Oncol. 2013;14(13):1269–77. doi: 10.1016/S1470-2045(13)70497-2. [DOI] [PubMed] [Google Scholar]
  • 29.Cuzick J SH, Peto R, et al. Overview of randomized trials of postoperative adjuvant radiotherapy in breast cancer. Cancer Treat Rep. 1987;71:15–29. [PubMed] [Google Scholar]
  • 30.Harness JK, Silverstein MJ, Wazer DE, et al. Radiotherapy for breast cancer, the TARGIT-A trial. Lancet. 2014;383(9930):1718–9. doi: 10.1016/S0140-6736(14)60829-1. [DOI] [PubMed] [Google Scholar]
  • 31.Mackenzie P, Fyles A, Chung C. Radiotherapy for breast cancer, the TARGIT-A trial. Lancet. 2014;383(9930):1717. doi: 10.1016/S0140-6736(14)60827-8. [DOI] [PubMed] [Google Scholar]
  • 32.Yarnold J, Offersen BV, Olivotto I, et al. Radiotherapy for breast cancer, the TARGIT-A trial. Lancet. 2014;383(9930):1717–8. doi: 10.1016/S0140-6736(14)60828-X. [DOI] [PubMed] [Google Scholar]
  • 33.Cuzick J. Radiotherapy for breast cancer, the TARGIT-A trial. Lancet. 2014;383(9930):1716. doi: 10.1016/S0140-6736(14)60825-4. [DOI] [PubMed] [Google Scholar]
  • 34.Haviland JS, A'Hern R, Bentzen SM, et al. Radiotherapy for breast cancer, the TARGIT-A trial. Lancet. 2014;383(9930):1716–7. doi: 10.1016/S0140-6736(14)60826-6. [DOI] [PubMed] [Google Scholar]
  • 35.Silverstein MJ, Fastner G, Maluta S, et al. Intraoperative radiation therapy: a critical analysis of the ELIOT and TARGIT trials Part 2–TARGIT. Ann Surg Oncol. 2014;21(12):3793–9. doi: 10.1245/s10434-014-3999-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Silverstein MJ, Fastner G, Maluta S, et al. Intraoperative radiation therapy: a critical analysis of the ELIOT and TARGIT trials Part 1–ELIOT. Ann Surg Oncol. 2014;21(12):3787–92. doi: 10.1245/s10434-014-3998-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Leonardi MC, Maisonneuve P, Mastropasqua MG, et al. Accelerated partial breast irradiation with intraoperative electrons: using GEC-ESTRO recommendations as guidance for patient selection. Radiother Oncol. 2013;106(1):21–7. doi: 10.1016/j.radonc.2012.10.018. [DOI] [PubMed] [Google Scholar]
  • 38.Leonardi MC, Maisonneuve P, Mastropasqua MG, et al. How do the ASTRO consensus statement guidelines for the application of accelerated partial breast irradiation fit intraoperative radiotherapy? A retrospective analysis of patients treated at the European Institute of Oncology. Int J Radiat Oncol Biol Phys. 2012;83(3):806–13. doi: 10.1016/j.ijrobp.2011.08.014. [DOI] [PubMed] [Google Scholar]
  • 39.Smith BD, Arthur DW, Buchholz TA, et al. Accelerated partial breast irradiation consensus statement from the American Society for Radiation Oncology (ASTRO) Int J Radiat Oncol Biol Phys. 2009;74(4):987–1001. doi: 10.1016/j.ijrobp.2009.02.031. [DOI] [PubMed] [Google Scholar]
  • 40.Polgar C, Van Limbergen E, Potter R, et al. Patient selection for accelerated partial-breast irradiation (APBI) after breast-conserving surgery: recommendations of the Groupe Europeen de Curietherapie-European Society for Therapeutic Radiology and Oncology (GEC-ESTRO) breast cancer working group based on clinical evidence (2009) Radiother Oncol. 2010;94(3):264–73. doi: 10.1016/j.radonc.2010.01.014. [DOI] [PubMed] [Google Scholar]
  • 41.Maluta S, Dall'Oglio S, Goer DA, et al. Intraoperative electron radiotherapy (IOERT) as an alternative to standard whole breast irradiation: only for low-risk subgroups? Breast care. 2014;9(2):102–6. doi: 10.1159/000362392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Fastner G, Sedlmayer F, et al. IORT with electrons as boost strategy during breast conserving therapy in limited stage breast cancer: long term results of an ISIORT pooled analysis. Radiother Oncol. 2013;108(2):279–86. doi: 10.1016/j.radonc.2013.05.031. [DOI] [PubMed] [Google Scholar]
  • 43.Vaidya JS, Baum M, Tobias JS, et al. Long-term results of targeted intraoperative radiotherapy (Targit) boost during breast-conserving surgery. Int J Radiat Oncol Biol Phys. 2011;81(4):1091–7. doi: 10.1016/j.ijrobp.2010.07.1996. [DOI] [PubMed] [Google Scholar]
  • 44.Blank E, Kraus-Tiefenbacher U, Welzel G, et al. Single-center long-term follow-up after intraoperative radiotherapy as a boost during breast-conserving surgery using low-kilovoltage x-rays. Ann Surg Oncol. 2010;17(Suppl 3):352–8. doi: 10.1245/s10434-010-1265-z. [DOI] [PubMed] [Google Scholar]
  • 45.Malter W, Puppe J, Rogee K, et al. Single center experiences with intraoperative radiotherapy as a boost during oncoplastic breast-conserving surgery. Eur J Cancer. 2012;48:S219. doi: 10.1016/S0959-8049(12)70663-9. [DOI] [Google Scholar]
  • 46.Malter W, Kirn V, Mallmann P, et al. Oncoplastic breast reconstruction after IORT. Transla Cancer Res. 2014;3(1):74–82. [Google Scholar]
  • 47.Braendengen M, Tveit KM, Berglund A, et al. Randomized phase III study comparing preoperative radiotherapy with chemoradiotherapy in nonresectable rectal cancer. J Clin Oncol. 2008;26(22):3687–94. doi: 10.1200/JCO.2007.15.3858. [DOI] [PubMed] [Google Scholar]
  • 48.Frykholm GJ, Pahlman L, Glimelius B. Combined chemo- and radiotherapy vs. radiotherapy alone in the treatment of primary, nonresectable adenocarcinoma of the rectum. Int J Radiat Oncol Biol Phys. 2001;50(2):427–34. doi: 10.1016/S0360-3016(01)01479-1. [DOI] [PubMed] [Google Scholar]
  • 49.Calvo FA, Gomez-Espi M, Diaz-Gonzalez JA, et al. Intraoperative presacral electron boost following preoperative chemoradiation in T3-4Nx rectal cancer: initial local effects and clinical outcome analysis. Radiother Oncol. 2002;62(2):201–6. doi: 10.1016/S0167-8140(01)00477-7. [DOI] [PubMed] [Google Scholar]
  • 50.Mathis KL, Nelson H, Pemberton JH, et al. Unresectable colorectal cancer can be cured with multimodality therapy. Ann Surg. 2008;248(4):592–8. doi: 10.1097/SLA.0b013e318187ed4a. [DOI] [PubMed] [Google Scholar]
  • 51.Minsky BD, Cohen AM, Enker WE, et al. Radiation therapy for unresectable rectal cancer. Int J Radiat Oncol Biol Phys. 21(5):1283–9. doi: 10.1016/0360-3016(91)90287-e. [DOI] [PubMed] [Google Scholar]
  • 52.Ratto C, Valentini V, Morganti AG, et al. Combined-modality therapy in locally advanced primary rectal cancer. Dis Colon Rectum. 2003;46(1):59–67. doi: 10.1007/s10350-004-6497-1. [DOI] [PubMed] [Google Scholar]
  • 53.Sadahiro S, Suzuki T, Ishikawa K, et al. Preoperative radio/chemo-radiotherapy in combination with intraoperative radiotherapy for T3-4Nx rectal cancer. Eur J Surg Oncol. 2004;30(7):750–8. doi: 10.1016/j.ejso.2004.04.012. [DOI] [PubMed] [Google Scholar]
  • 54.Ferenschild FT, Vermaas M, Nuyttens JJ, et al. Value of intraoperative radiotherapy in locally advanced rectal cancer. Dis Colon Rectum. 2006;49(9):1257–65. doi: 10.1007/s10350-006-0651-x. [DOI] [PubMed] [Google Scholar]
  • 55.Roeder F, Treiber M, Oertel S, et al. Patterns of failure and local control after intraoperative electron boost radiotherapy to the presacral space in combination with total mesorectal excision in patients with locally advanced rectal cancer. Int J Radiat Oncol Biol Phys. 2007;67(5):1381–8. doi: 10.1016/j.ijrobp.2006.11.039. [DOI] [PubMed] [Google Scholar]
  • 56.Masaki T, Takayama M, Matsuoka H, et al. Intraoperative radiotherapy for oncological and function-preserving surgery in patients with advanced lower rectal cancer. Langenbecks Arch Surg. 2008;393(2):173–80. doi: 10.1007/s00423-007-0260-8. [DOI] [PubMed] [Google Scholar]
  • 57.Valentini V, Coco C, Rizzo G, et al. Outcomes of clinical T4M0 extra-peritoneal rectal cancer treated with preoperative radiochemotherapy and surgery: a prospective evaluation of a single institutional experience. Surgery. 2009;145(5):486–94. doi: 10.1016/j.surg.2009.01.007. [DOI] [PubMed] [Google Scholar]
  • 58.Dubois JB, Bussieres E, Richaud P, et al. Intra-operative radiotherapy of rectal cancer: results of the French multi-institutional randomized study. Radiother Oncol. 2011;98(3):298–303. doi: 10.1016/j.radonc.2011.01.017. [DOI] [PubMed] [Google Scholar]
  • 59.Kusters M, Valentini V, Calvo FA, et al. Results of European pooled analysis of IORT-containing multimodality treatment for locally advanced rectal cancer: adjuvant chemotherapy prevents local recurrence rather than distant metastases. Ann Oncol. 2010;21(6):1279–84. doi: 10.1093/annonc/mdp501. [DOI] [PubMed] [Google Scholar]
  • 60.Sole CV, Calvo FA, Serrano J, et al. Post-chemoradiation intraoperative electron-beam radiation therapy boost in resected locally advanced rectal cancer: long-term results focused on topographic pattern of locoregional relapse. Radiother Oncol. 2014;112(1):52–8. doi: 10.1016/j.radonc.2014.05.012. [DOI] [PubMed] [Google Scholar]
  • 61.Holman FA, Haddock MG, Gunderson LL, et al. Results of intraoperative electron beam radiotherapy containing multimodality treatment for locally unresectable T4 rectal cancer: a pooled analysis of the Mayo Clinic Rochester and Catharina Hospital Eindhoven. J Gastrointest Oncol. 2016;7(6):903–16. doi: 10.21037/jgo.2016.07.01. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Willett CG, Shellito PC, Tepper JE, et al. Intraoperative electron beam radiation therapy for primary locally advanced rectal and rectosigmoid carcinoma. J Clin Oncol. 1991;9(5):843–9. doi: 10.1200/JCO.1991.9.5.843. [DOI] [PubMed] [Google Scholar]
  • 63.Suzuki K, Gunderson LL, Devine RM, et al. Intraoperative irradiation after palliative surgery for locally recurrent rectal cancer. Cancer. 1995;75(4):939–52. doi: 10.1002/1097-0142(19950215)75:4<939::aid-cncr2820750408>3.0.co;2-e. [DOI] [PubMed] [Google Scholar]
  • 64.Willett CG, Shellito PC, Tepper JE, et al. Intraoperative electron beam radiation therapy for recurrent locally advanced rectal or rectosigmoid carcinoma. Cancer. 1991;67(6):1504–8. doi: 10.1002/1097-0142(19910315)67:6<1504::aid-cncr2820670607>3.0.co;2-x. [DOI] [PubMed] [Google Scholar]
  • 65.Haddock MG, Miller RC, Nelson H, et al. Combined modality therapy including intraoperative electron irradiation for locally recurrent colorectal cancer. Int J Radiat Oncol Biol Phys. 2011;79(1):143–50. doi: 10.1016/j.ijrobp.2009.10.046. [DOI] [PubMed] [Google Scholar]
  • 66.Valentini V, Morganti AG, De Franco A, et al. Chemoradiation with or without intraoperative radiation therapy in patients with locally recurrent rectal carcinoma: prognostic factors and long term outcome. Cancer. 1999;86(12):2612–24. doi: 10.1002/(sici)1097-0142(19991215)86:12<2612::aid-cncr5>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
  • 67.Wiig JN, Tveit KM, Poulsen JP, et al. Preoperative irradiation and surgery for recurrent rectal cancer. Will intraoperative radiotherapy (IORT) be of additional benefit? A prospective study. Radiother Oncol. 2002;62(2):207–13. doi: 10.1016/S0167-8140(01)00486-8. [DOI] [PubMed] [Google Scholar]
  • 68.Calvo FA, Sole CV, Alvarez de Sierra P, et al. Prognostic impact of external beam radiation therapy in patients treated with and without extended surgery and intraoperative electrons for locally recurrent rectal cancer: 16-year experience in a single institution. Int J Radiat Oncol Biol Phys. 2013;86(5):892–900. doi: 10.1016/j.ijrobp.2013.04.008. [DOI] [PubMed] [Google Scholar]
  • 69.Dresen RC, Gosens MJ, Martijn H, et al. Radical resection after IORT-containing multimodality treatment is the most important determinant for outcome in patients treated for locally recurrent rectal cancer. Ann Surg Oncol. 2008;15(7):1937–47. doi: 10.1245/s10434-008-9896-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Roeder F, Goetz JM, Habl G, et al. Intraoperative electron radiation therapy (IOERT) in the management of locally recurrent rectal cancer. BMC Cancer. 2012;12:592. doi: 10.1186/1471-2407-12-592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Holman FA, Bosman SJ, Haddock MG, et al. Results of a pooled analysis of IOERT containing multimodality treatment for locally recurrent rectal cancer: results of 565 patients of two major treatment centres. Eur J Surg Oncol. 2017;43(1):107–17. doi: 10.1016/j.ejso.2016.08.015. [DOI] [PubMed] [Google Scholar]
  • 72.Huber FT, Stepan R, Zimmermann F, et al. Locally advanced rectal cancer: resection and intraoperative radiotherapy using the flab method combined with preoperative or postoperative radiochemotherapy. Dis Colon Rectum. 1996;39(7):774–9. doi: 10.1007/BF02054443. [DOI] [PubMed] [Google Scholar]
  • 73.Nuyttens JJ, Kolkman-Deurloo IK, Vermaas M, et al. High-dose-rate intraoperative radiotherapy for close or positive margins in patients with locally advanced or recurrent rectal cancer. Int J Radiat Oncol Biol Phys. 2004;58(1):106–12. doi: 10.1016/S0360-3016(03)01494-9. [DOI] [PubMed] [Google Scholar]
  • 74.Mirnezami R, Chang GJ, Das P, et al. Intraoperative radiotherapy in colorectal cancer: systematic review and meta-analysis of techniques, long-term outcomes, and complications. Surg Oncol. 2013;22(1):22–35. doi: 10.1016/j.suronc.2012.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Alektiar KM, Zelefsky MJ, Paty PB, et al. High-dose-rate intraoperative brachytherapy for recurrent colorectal cancer. Int J Radiat Oncol Biol Phys. 2000;48(1):219–26. doi: 10.1016/S0360-3016(00)00634-9. [DOI] [PubMed] [Google Scholar]
  • 76.Lindel K, Willett CG, Shellito PC, et al. Intraoperative radiation therapy for locally advanced recurrent rectal or rectosigmoid cancer. Radiother Oncol. 2001;58(1):83–7. doi: 10.1016/S0167-8140(00)00309-1. [DOI] [PubMed] [Google Scholar]
  • 77.Edmonson JH, Petersen IA, Shives TC, et al. Chemotherapy, irradiation, and surgery for function-preserving therapy of primary extremity soft tissue sarcomas: initial treatment with ifosfamide, mitomycin, doxorubicin, and cisplatin plus granulocyte macrophage-colony-stimulating factor. Cancer. 2002;94(3):786–92. doi: 10.1002/cncr.10259. [DOI] [PubMed] [Google Scholar]
  • 78.Azinovic I, Martinez Monge R, Aristu JJ, et al. Intraoperative radiotherapy electron boost followed by moderate doses of external beam radiotherapy in resected soft-tissue sarcoma of the extremities. Radiother Oncol. 2003;67(3):331–7. doi: 10.1016/S0167-8140(03)00163-4. [DOI] [PubMed] [Google Scholar]
  • 79.Kretzler A, Molls M, Gradinger R, et al. Intraoperative radiotherapy of soft tissue sarcoma of the extremity. Strahlenther Onkol. 2004;180(6):365–70. doi: 10.1007/s00066-004-1191-8. [DOI] [PubMed] [Google Scholar]
  • 80.Oertel S, Treiber M, Zahlten-Hinguranage A, et al. Intraoperative electron boost radiation followed by moderate doses of external beam radiotherapy in limb-sparing treatment of patients with extremity soft-tissue sarcoma. Int J Radiat Oncol Biol Phys. 2006;64(5):1416–23. doi: 10.1016/j.ijrobp.2005.10.009. [DOI] [PubMed] [Google Scholar]
  • 81.Niewald M, Fleckenstein J, Licht N, et al. Intraoperative radiotherapy (IORT) combined with external beam radiotherapy (EBRT) for soft-tissue sarcomas—a retrospective evaluation of the Homburg experience in the years 1995–2007. Radiat Oncol. 2009;4:32. doi: 10.1186/1748-717X-4-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Call JA, Stafford SL, Petersen IA, et al. Use of intraoperative radiotherapy for upper-extremity soft-tissue sarcomas: analysis of disease outcomes and toxicity. Am J Clin Oncol. 2014;37(1):81–5. doi: 10.1097/COC.0b013e31826b9b3d. [DOI] [PubMed] [Google Scholar]
  • 83.Calvo FA, Sole CV, Polo A, et al. Limb-sparing management with surgical resection, external-beam and intraoperative electron-beam radiation therapy boost for patients with primary soft tissue sarcoma of the extremity: a multicentric pooled analysis of long-term outcomes. Strahlenther Onkol. 2014;190(10):891–8. doi: 10.1007/s00066-014-0640-2. [DOI] [PubMed] [Google Scholar]
  • 84.Roeder F, Lehner B, Schmitt T, et al. Excellent local control with IOERT and postoperative EBRT in high grade extremity sarcoma: results from a subgroup analysis of a prospective trial. BMC Cancer. 2014;14:350. doi: 10.1186/1471-2407-14-350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Roeder F, Lehner B, Saleh-Ebrahimi L, et al. Intraoperative electron radiation therapy combined with external beam radiation therapy and limb sparing surgery in extremity soft tissue sarcoma: a retrospective single center analysis of 183 cases. Radiother Oncol. 2016;119(1):22–9. doi: 10.1016/j.radonc.2015.11.014. [DOI] [PubMed] [Google Scholar]
  • 86.Sindelar WF, Kinsella TJ, Chen PW, et al. Intraoperative radiotherapy in retroperitoneal sarcomas. Final results of a prospective, randomized, clinical trial. Arch Surg. 1993;128(4):402–10. doi: 10.1001/archsurg.1993.01420160040005. [DOI] [PubMed] [Google Scholar]
  • 87.Alektiar KM, Hu K, Anderson L, et al. High-dose-rate intraoperative radiation therapy (HDR-IORT) for retroperitoneal sarcomas. Int J Radiat Oncol Biol Phys. 2000;47(1):157–63. doi: 10.1016/S0360-3016(99)00546-5. [DOI] [PubMed] [Google Scholar]
  • 88.Gieschen HL, Spiro IJ, Suit HD, et al. Long-term results of intraoperative electron beam radiotherapy for primary and recurrent retroperitoneal soft tissue sarcoma. Int J Radiat Oncol Biol Phys. 2001;50(1):127–31. doi: 10.1016/S0360-3016(00)01589-3. [DOI] [PubMed] [Google Scholar]
  • 89.Petersen IA, Haddock MG, Donohue JH, et al. Use of intraoperative electron beam radiotherapy in the management of retroperitoneal soft tissue sarcomas. Int J Radiat Oncol Biol Phys. 2002;52(2):469–75. doi: 10.1016/S0360-3016(01)02595-0. [DOI] [PubMed] [Google Scholar]
  • 90.Bobin JY, Al-Lawati T, Granero LE, et al. Surgical management of retroperitoneal sarcomas associated with external and intraoperative electron beam radiotherapy. Eur J Surg Oncol. 2003;29(8):676–81. doi: 10.1016/S0748-7983(03)00139-2. [DOI] [PubMed] [Google Scholar]
  • 91.Pierie JP, Betensky RA, Choudry U, et al. Outcomes in a series of 103 retroperitoneal sarcomas. Eur J Surg Oncol. 2006;32(10):1235–41. doi: 10.1016/j.ejso.2006.07.002. [DOI] [PubMed] [Google Scholar]
  • 92.Krempien R, Roeder F, Oertel S, et al. Intraoperative electron-beam therapy for primary and recurrent retroperitoneal soft-tissue sarcoma. Int J Radiat Oncol Biol Phys. 2006;65(3):773–9. doi: 10.1016/j.ijrobp.2006.01.028. [DOI] [PubMed] [Google Scholar]
  • 93.Pawlik TM, Pisters PW, Mikula L, et al. Long-term results of two prospective trials of preoperative external beam radiotherapy for localized intermediate- or high-grade retroperitoneal soft tissue sarcoma. Ann Surg Oncol. 2006;13(4):508–17. doi: 10.1245/ASO.2006.05.035. [DOI] [PubMed] [Google Scholar]
  • 94.Ballo MT, Zagars GK, Pollock RE, et al. Retroperitoneal soft tissue sarcoma: an analysis of radiation and surgical treatment. Int J Radiat Oncol Biol Phys. 2007;67(1):158–63. doi: 10.1016/j.ijrobp.2006.08.025. [DOI] [PubMed] [Google Scholar]
  • 95.Dziewirski W, Rutkowski P, Nowecki ZI, et al. Surgery combined with intraoperative brachytherapy in the treatment of retroperitoneal sarcomas. Ann Surg Oncol. 2006;13(2):245–52. doi: 10.1245/ASO.2006.03.026. [DOI] [PubMed] [Google Scholar]
  • 96.Sweeting RS, Deal AM, Llaguna OH, et al. Intraoperative electron radiation therapy as an important treatment modality in retroperitoneal sarcoma. J Surg Res. 2013;185(1):245–9. doi: 10.1016/j.jss.2013.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Roeder F, Ulrich A, Habl G, et al. Clinical phase I/II trial to investigate preoperative dose-escalated intensity-modulated radiation therapy (IMRT) and intraoperative radiation therapy (IORT) in patients with retroperitoneal soft tissue sarcoma: interim analysis. BMC Cancer. 2014;14:617. doi: 10.1186/1471-2407-14-617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Stucky CC, Wasif N, Ashman JB, et al. Excellent local control with preoperative radiation therapy, surgical resection, and intra-operative electron radiation therapy for retroperitoneal sarcoma. J Surg Oncol. 2014;109(8):798–803. doi: 10.1002/jso.23576. [DOI] [PubMed] [Google Scholar]
  • 99.Gronchi A, De Paoli A, Dani C, et al. Preoperative chemo-radiation therapy for localised retroperitoneal sarcoma: a phase I-II study from the Italian Sarcoma Group. Eur J Cancer. 2014;50(4):784–92. doi: 10.1016/j.ejca.2013.11.021. [DOI] [PubMed] [Google Scholar]
  • 100.Miller RC, Haddock MG, Petersen IA, et al. Intraoperative electron-beam radiotherapy and ureteral obstruction. Int J Radiat Oncol Biol Phys. 2006;64(3):792–8. doi: 10.1016/j.ijrobp.2005.08.019. [DOI] [PubMed] [Google Scholar]
  • 101.Calvo FA, Sole CV, Cambeiro M, et al. Prognostic value of external beam radiation therapy in patients treated with surgical resection and intraoperative electron beam radiation therapy for locally recurrent soft tissue sarcoma: a multicentric long-term outcome analysis. Int J Radiat Oncol Biol Phys. 2014;88(1):143–50. doi: 10.1016/j.ijrobp.2013.10.021. [DOI] [PubMed] [Google Scholar]
  • 102.Haase GM, Meagher DP, Jr., McNeely LK, et al. Electron beam intraoperative radiation therapy for pediatric neoplasms. Cancer. 1994;74(2):740–7. doi: 10.1002/1097-0142(19940715)74:2<740::aid-cncr2820740230>3.0.co;2-7. [DOI] [PubMed] [Google Scholar]
  • 103.Oertel S, Niethammer AG, Krempien R, et al. Combination of external-beam radiotherapy with intraoperative electron-beam therapy is effective in incompletely resected pediatric malignancies. Int J Radiat Oncol Biol Phys. 2006;64(1):235–41. doi: 10.1016/j.ijrobp.2005.06.038. [DOI] [PubMed] [Google Scholar]
  • 104.Goodman KA, Wolden SL, LaQuaglia MP, et al. Intraoperative high-dose-rate brachytherapy for pediatric solid tumors: a 10-year experience. Brachytherapy. 2003;2(3):139–46. doi: 10.1016/S1538-4721(03)00135-1. [DOI] [PubMed] [Google Scholar]
  • 105.Sole CV, Calvo FA, Polo A, et al. Intraoperative electron-beam radiation therapy for pediatric Ewing sarcomas and rhabdomyosarcomas: long-term outcomes. Int J Radiat Oncol Biol, Phys. 2015;92(5):1069–76. doi: 10.1016/j.ijrobp.2015.04.048. [DOI] [PubMed] [Google Scholar]
  • 106.Nag S, Tippin D, Smith S, et al. Intraoperative electron beam treatment for pediatric malignancies: The Ohio State University experience. Med Pediatr Oncol. 2003;40(6):360–6. doi: 10.1002/mpo.10296. [DOI] [PubMed] [Google Scholar]
  • 107.Stauder MC, Laack NN, Moir CR, et al. Excellent local control and survival after intraoperative and external beam radiotherapy for pediatric solid tumors: long-term follow-up of the Mayo Clinic experience. J Pediatr Hematol/Oncol. 2011;33(5):350–5. doi: 10.1097/MPH.0b013e3182148dad. [DOI] [PubMed] [Google Scholar]
  • 108.Brunschwig A, Barber HR. Extended pelvic exenteration for advanced cancer of the cervix. long survivals following added resection of involved small bowel. Cancer. 1964;17:1267–70. doi: 10.1002/1097-0142(196410)17:10<1267::aid-cncr2820171008>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
  • 109.Karlen JR, Piver MS. Reduction of mortality and morbidity associated with pelvic exenteration. Gynecol Oncol. 1975;3(2):164–7. doi: 10.1016/0090-8258(75)90076-1. [DOI] [PubMed] [Google Scholar]
  • 110.Kiselow M, Butcher HR, Jr., Bricker EM. Results of the radical surgical treatment of advanced pelvic cancer: a fifteen-year study. Ann Surg. 1967;166(3):428–36. [PMC free article] [PubMed] [Google Scholar]
  • 111.Symmonds RE, Pratt JH, Webb MJ. Exenterative operations: experience with 198 patients. Am J Obstet Gynecol. 1975;121(7):907–18. doi: 10.1016/0002-9378(75)90908-4. [DOI] [PubMed] [Google Scholar]
  • 112.Giorda G, Boz G, Gadducci A, et al. Multimodality approach in extra cervical locally advanced cervical cancer: chemoradiation, surgery and intra-operative radiation therapy A phase II trial. Eur J Surg Oncol. 2011;37(5):442–7. doi: 10.1016/j.ejso.2011.02.011. [DOI] [PubMed] [Google Scholar]
  • 113.Garton GR, Gunderson LL, Webb MJ, et al. Intraoperative radiation therapy in gynecologic cancer: update of the experience at a single institution. Int J Radiat Oncol Biol Phys. 1997;37(4):839–43. doi: 10.1016/S0360-3016(96)00546-9. [DOI] [PubMed] [Google Scholar]
  • 114.Gemignani ML, Alektiar KM, Leitao M, et al. Radical surgical resection and high-dose intraoperative radiation therapy (HDR-IORT) in patients with recurrent gynecologic cancers. Int J Radiat Oncol Biol Phys. 2001;50(3):687–94. doi: 10.1016/S0360-3016(01)01507-3. [DOI] [PubMed] [Google Scholar]
  • 115.Haddock MG, Petersen IA, Webb MJ, et al. IORT for locally advanced gynecological malignancies. Front Radiat Ther Oncol. 1997;31:256–9. doi: 10.1159/000061131. [DOI] [PubMed] [Google Scholar]
  • 116.Martinez-Monge R, Jurado M, et al. Intraoperative electron beam radiotherapy during radical surgery for locally advanced and recurrent cervical cancer. Gynecol Oncol. 2001;82(3):538–43. doi: 10.1006/gyno.2001.6329. [DOI] [PubMed] [Google Scholar]
  • 117.Sole CV, Calvo FA, Lozano MA, et al. External-beam radiation therapy after surgical resection and intraoperative electron-beam radiation therapy for oligorecurrent gynecological cancer Long-term outcome. Strahlenther Onkol. 2014;190(2):171–80. doi: 10.1007/s00066-013-0472-5. [DOI] [PubMed] [Google Scholar]
  • 118.Tran PT, Su Z, Hara W, et al. Long-term survivors using intraoperative radiotherapy for recurrent gynecologic malignancies. Int J Radiat Oncol Biol Phys. 2007;69(2):504–11. doi: 10.1016/j.ijrobp.2007.03.021. [DOI] [PubMed] [Google Scholar]
  • 119.Barney BM, Petersen IA, Dowdy SC, et al. Intraoperative electron beam radiotherapy (IOERT) in the management of locally advanced or recurrent cervical cancer. Radiat Oncol. 2013;8:80. doi: 10.1186/1748-717X-8-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Calvo FA, Sole CV, Lozano MA, et al. Intraoperative electron beam radiotherapy and extended surgical resection for gynecological pelvic recurrent malignancies with and without external beam radiation therapy: long-term outcomes. Gynecol Oncol. 2013;130(3):537–44. doi: 10.1016/j.ygyno.2013.05.016. [DOI] [PubMed] [Google Scholar]
  • 121.Foley OW, Rauh-Hain JA, Clark RM, et al. Intraoperative radiation therapy in the management of gynecologic malignancies. Am J Clin Oncol. 2016;39(4):329–34. doi: 10.1097/COC.0000000000000063. [DOI] [PubMed] [Google Scholar]
  • 122.Arians N, Foerster R, Rom J, et al. Outcome of patients with local recurrent gynecologic malignancies after resection combined with intraoperative electron radiation therapy (IOERT) Radiat Oncol. 2016;11:44. doi: 10.1186/s13014-016-0622-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Dowdy SC, Mariani A, Cliby WA, et al. Radical pelvic resection and intraoperative radiation therapy for recurrent endometrial cancer: technique and analysis of outcomes. Gynecol Oncol. 2006;101(2):280–6. doi: 10.1016/j.ygyno.2005.10.018. [DOI] [PubMed] [Google Scholar]
  • 124.Mahe MA, Gerard JP, Dubois JB, et al. Intraoperative radiation therapy in recurrent carcinoma of the uterine cervix: report of the French intraoperative group on 70 patients. Int J Radiat Oncol Biol Phys. 1996;34(1):21–6. doi: 10.1016/0360-3016(95)02089-6. [DOI] [PubMed] [Google Scholar]
  • 125.Mahe MA, Romestaing P, Gerard JP, et al. Prognostic factors for local control in recurrent cervical carcinoma treated with IORT: report of the French IORT Group. Front Radiat Ther Oncol. 1997;31:267–70. doi: 10.1159/000061190. [DOI] [PubMed] [Google Scholar]
  • 126.del Carmen MG, McIntyre JF, Fuller AF, et al. Intraoperative radiation therapy in the treatment of pelvic gynecologic malignancies: a review of fifteen cases. Gynecol Oncol. 2000;79(3):457–62. doi: 10.1006/gyno.2000.6002. [DOI] [PubMed] [Google Scholar]
  • 127.Backes FJ, Billingsley CC, Martin DD, et al. Does intra-operative radiation at the time of pelvic exenteration improve survival for patients with recurrent, previously irradiated cervical, vaginal, or vulvar cancer? Gynecol Oncol. 2014;135(1):95–9. doi: 10.1016/j.ygyno.2014.07.093. [DOI] [PubMed] [Google Scholar]
  • 128.Hallemeier CL, Karnes RJ, Pisansky TM, et al. Multimodality therapy including surgical resection and intraoperative electron radiotherapy for recurrent or advanced primary carcinoma of the urinary bladder or ureter. Am J Clin Oncol. 2013;36(6):596–600. doi: 10.1097/COC.0b013e31825d52f7. [DOI] [PubMed] [Google Scholar]
  • 129.Hallemeier CL, Choo R, Davis BJ, et al. Long-term outcomes after maximal surgical resection and intraoperative electron radiotherapy for locoregionally recurrent or locoregionally advanced primary renal cell carcinoma. Int J Radiat Oncol Biol Phys. 2012:82. doi: 10.1016/j.ijrobp.2011.02.026. [DOI] [PubMed] [Google Scholar]
  • 130.Calvo FA, Sole CV, Martinez-Monge R, et al. Intraoperative EBRT and resection for renal cell carcinoma: twenty-year outcomes. Strahlentherapie Onkol. 2013;189 doi: 10.1007/s00066-012-0272-3. [DOI] [PubMed] [Google Scholar]
  • 131.Eble MJ, Staehler G, Wannenmacher M. The intraoperative radiotherapy (IORT) of locally spread and recurrent renal-cell carcinomas. Strahlentherapie Onkol. 1998;174(1):30–6. doi: 10.1007/BF03038225. [DOI] [PubMed] [Google Scholar]
  • 132.Habl G, Uhl M, Hensley F, et al. Intraoperative electron radiation therapy (IOERT) in patients with locally recurrent renal cell carcinoma. Radiat Oncol. 2013;8(1):282. doi: 10.1186/1748-717X-8-282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Master VA, Gottschalk AR, Kane C, et al. Management of isolated renal fossa recurrence following radical nephrectomy. J Urol. 2005;174 doi: 10.1097/01.ju.0000165574.62188.d0. [DOI] [PubMed] [Google Scholar]
  • 134.Paly JJ, Hallemeier CL, Biggs PJ, et al. Outcomes for a multi-institutional cohort of patients treated with intraoperative radiation therapy for advanced or recurrent renal cell carcinoma [abstract] Int J Oncol Biol Phys. 2012;84 doi: 10.1016/j.ijrobp.2012.07.1123. [DOI] [PubMed] [Google Scholar]
  • 135.Thompson IM, Valicenti RK, Albertsen P, et al. Adjuvant and salvage radiotherapy after prostatectomy: AUA/ASTRO Guideline. J Urol. 2013;190(2):441–9. doi: 10.1016/j.juro.2013.05.032. [DOI] [PubMed] [Google Scholar]
  • 136.Rocco B, Jereczek-Fossa BA, Matei DV, et al. Intraoperative radiotherapy during radical prostatectomy for intermediate-risk to locally advanced prostate cancer: treatment technique and evaluation of perioperative and functional outcome vs standard radical prostatectomy, in a matched-pair analysis. BJU Int. 2009;104(11):1624–30. doi: 10.1111/j.1464-410X.2009.08668.x. [DOI] [PubMed] [Google Scholar]
  • 137.Higashi Y, Hyochi N, Tari K. Intraoperative radiotherapy combined with external beam radiation for prostate cancer without metastasis. Nihon Rinsho. 1998;56(8):2177–80. [PubMed] [Google Scholar]
  • 138.Kato S, Sakura M, Kazumoto T, et al. Intraoperative radiation therapy for locally advanced prostate cancer. J JASTRO. 1998;10(3):241–8. [Google Scholar]
  • 139.Krengli M, Terrone C, Ballare A, et al. Intraoperative radiotherapy during radical prostatectomy for locally advanced prostate cancer: technical and dosimetric aspects. Int J Radiat Oncol Biol Phys. 2010;76(4):1073–7. doi: 10.1016/j.ijrobp.2009.03.037. [DOI] [PubMed] [Google Scholar]
  • 140.Orecchia R, Jereczek-Fossa BA, Ciocca M, et al. Intraoperative radiotherapy for locally advanced prostate cancer: treatment technique and ultrasound-based analysis of dose distribution. Anticancer Res. 2007;27(5b):3471–6. [PubMed] [Google Scholar]
  • 141.Saracino B, Gallucci M, et al. Phase I-II study of intraoperative radiation therapy (IORT) after radical prostatectomy for prostate cancer. Int J Radiat Oncol Biol Phys. 2008;71(4):1049–56. doi: 10.1016/j.ijrobp.2007.11.076. [DOI] [PubMed] [Google Scholar]
  • 142.Wisbeck WM, Becher EM, Russell AH. Adenocarcinoma of the stomach: autopsy observations with therapeutic implications for the radiation oncologist. Radiother Oncol. 1986;7(1):13–8. doi: 10.1016/S0167-8140(86)80120-7. [DOI] [PubMed] [Google Scholar]
  • 143.Gunderson LL. Gastric cancer—patterns of relapse after surgical resection. Semin Radiat Oncol. 2002;12(2):150–61. doi: 10.1053/srao.2002.30817. [DOI] [PubMed] [Google Scholar]
  • 144.Macdonald JS, Smalley SR, Benedetti J, et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med. 2001;345(10):725–30. doi: 10.1056/NEJMoa010187. [DOI] [PubMed] [Google Scholar]
  • 145.Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. New Engl J Med. 2006;355(1):11–20. doi: 10.1056/NEJMoa055531. [DOI] [PubMed] [Google Scholar]
  • 146.Wu CW, Hsiung CA, Lo SS, et al. Nodal dissection for patients with gastric cancer: a randomised controlled trial. Lancet Oncol. 2006;7(4):309–15. doi: 10.1016/S1470-2045(06)70623-4. [DOI] [PubMed] [Google Scholar]
  • 147.Maruyama K, Sasako M, Kinoshita T, et al. Pancreas-preserving total gastrectomy for proximal gastric cancer. World J Surg. 1995;19(4):532–6. doi: 10.1007/BF00294714. [DOI] [PubMed] [Google Scholar]
  • 148.Fu S, Lu JJ, Zhang Q, et al. Intraoperative radiotherapy combined with adjuvant chemoradiotherapy for locally advanced gastric adenocarcinoma. Int J Radiat Oncol, Biol, Phys. 2008;72(5):1488–94. doi: 10.1016/j.ijrobp.2008.03.012. [DOI] [PubMed] [Google Scholar]
  • 149.Sindelar WF, Kinsella TJ, Tepper JE, et al. Randomized trial of intraoperative radiotherapy in carcinoma of the stomach. Am J Surg. 1993;165(1):178–86. doi: 10.1016/S0002-9610(05)80423-4. [DOI] [PubMed] [Google Scholar]
  • 150.Zhang Q, Tey J, Peng L, et al. Adjuvant chemoradiotherapy with or without intraoperative radiotherapy for the treatment of resectable locally advanced gastric adenocarcinoma. Radiother Oncol. 2012;102(1):51–5. doi: 10.1016/j.radonc.2011.10.008. [DOI] [PubMed] [Google Scholar]
  • 151.Drognitz O, Henne K, Weissenberger C, et al. Long-term results after intraoperative radiation therapy for gastric cancer. Int J Radiat Oncol Biol Phys. 2008;70(3):715–21. doi: 10.1016/j.ijrobp.2007.07.2331. [DOI] [PubMed] [Google Scholar]
  • 152.Ogata T, Araki K, Matsuura K, et al. A 10-year experience of intraoperative radiotherapy for gastric carcinoma and a new surgical method of creating a wider irradiation field for cases of total gastrectomy patients. Int J Radiat Oncol Biol Phys. 1995;32(2):341–7. doi: 10.1016/0360-3016(94)00479-5. [DOI] [PubMed] [Google Scholar]
  • 153.Qin HL, Lin CH, Zhang XL. Evaluation of intraoperative radiotherapy for gastric carcinoma with D2 and D3 surgical resection. World J Gastroenterol. 2006;12(43):7033–7. doi: 10.3748/wjg.v12.i43.7033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Skoropad VY, Berdov BA, Mardynski YS, et al. A prospective, randomized trial of pre-operative and intraoperative radiotherapy versus surgery alone in resectable gastric cancer. Eur J Surg Oncol. 2000;26(8):773–9. doi: 10.1053/ejso.2000.1002. [DOI] [PubMed] [Google Scholar]
  • 155.Calvo FA, Sole CV, Obregon R, et al. Intraoperative radiotherapy for the treatment of resectable locally advanced gastric adenocarcinoma: topography of locoregional recurrences and long-term outcomes. Clin Transl Oncol. 2013;15(6):443–9. doi: 10.1007/s12094-012-0949-1. [DOI] [PubMed] [Google Scholar]
  • 156.Coquard R, Ayzac L, Gilly FN, et al. Intraoperative radiation therapy combined with limited lymph node resection in gastric cancer: an alternative to extended dissection? Int J Radiat Oncol Biol Phys. 1997;39(5):1093–8. doi: 10.1016/S0360-3016(97)00386-6. [DOI] [PubMed] [Google Scholar]
  • 157.Bacalbasa N, Balescu I, Calin M, et al. Intraoperative radiation therapy in gastric cancer. J Med life. 2014;7(2):128–31. [PMC free article] [PubMed] [Google Scholar]
  • 158.Avizonis VN, Buzydlowski J, Lanciano R, et al. Treatment of adenocarcinoma of the stomach with resection, intraoperative radiotherapy, and adjuvant external beam radiation: a phase II study from Radiation Therapy Oncology Group 85-04. Ann Surg Oncol. 1995;2(4):295–302. doi: 10.1007/BF02307060. [DOI] [PubMed] [Google Scholar]
  • 159.Weese JL, Harbison SP, Stiller GD, et al. Neoadjuvant chemotherapy, radical resection with intraoperative radiation therapy (IORT): improved treatment for gastric adenocarcinoma. Surgery. 2000;128(4):564–71. doi: 10.1067/msy.2000.108420. [DOI] [PubMed] [Google Scholar]
  • 160.Glehen O, Peyrat P, Beaujard AC, et al. Pattern of failures in gastric cancer patients with lymph node involvement treated by surgery, intraoperative and external beam radiotherapy. Radiother Oncol. 2003;67(2):171–5. doi: 10.1016/S0167-8140(02)00344-4. [DOI] [PubMed] [Google Scholar]
  • 161.Miller RC, Haddock MG, Gunderson LL, et al. Intraoperative radiotherapy for treatment of locally advanced and recurrent esophageal and gastric adenocarcinomas. Dis Esophagus. 2006;19(6):487–95.. doi: 10.1111/j.1442-2050.2006.00626.x. [DOI] [PubMed] [Google Scholar]
  • 162.Gourgou-Bourgade S, Bascoul-Mollevi C, Desseigne F, et al. Impact of FOLFIRINOX compared with gemcitabine on quality of life in patients with metastatic pancreatic cancer: results from the PRODIGE 4/ACCORD 11 randomized trial. J Clin Oncol. 2013;31(1):23–9. doi: 10.1200/JCO.2012.44.4869. [DOI] [PubMed] [Google Scholar]
  • 163.Hazard L, Tward JD, Szabo A, et al. Radiation therapy is associated with improved survival in patients with pancreatic adenocarcinoma: results of a study from the Surveillance, Epidemiology, and End Results (SEER) registry data. Cancer. 2007;110(10):2191–201. doi: 10.1002/cncr.23047. [DOI] [PubMed] [Google Scholar]
  • 164.Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–703. doi: 10.1056/NEJMoa1304369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Alfieri S, Morganti AG, Di Giorgio A, et al. Improved survival and local control after intraoperative radiation therapy and postoperative radiotherapy: a multivariate analysis of 46 patients undergoing surgery for pancreatic head cancer. Arch Surg. 2001;136(3):343–7. doi: 10.1001/archsurg.136.3.343. [DOI] [PubMed] [Google Scholar]
  • 166.Calvo FA, Sole CV, Atahualpa F, et al. Chemoradiation for resected pancreatic adenocarcinoma with or without intraoperative radiation therapy boost: long-term outcomes. Pancreatology. 2013;13(6):576–82. doi: 10.1016/j.pan.2013.09.002. [DOI] [PubMed] [Google Scholar]
  • 167.Showalter TN, Rao AS, Anne PR, et al. Does intraoperative radiation therapy improve local tumor control in patients undergoing pancreaticoduodenectomy for pancreatic adenocarcinoma? A propensity score analysis. Ann Surg Oncol. 2009;16(8):2116–22. doi: 10.1245/s10434-009-0498-1. [DOI] [PubMed] [Google Scholar]
  • 168.Valentini V, Calvo F, Reni M, et al. Intra-operative radiotherapy (IORT) in pancreatic cancer: joint analysis of the ISIORT-Europe experience. Radiother Oncol. 2009;91(1):54–9. doi: 10.1016/j.radonc.2008.07.020. [DOI] [PubMed] [Google Scholar]
  • 169.Messick C, Hardacre JM, McGee MF, et al. Early experience with intraoperative radiotherapy in patients with resected pancreatic adenocarcinoma. Am J Surg. 2008;195(3):308–11. doi: 10.1016/j.amjsurg.2007.12.024. [DOI] [PubMed] [Google Scholar]
  • 170.Ogawa K, Karasawa K, Ito Y, et al. Intraoperative radiotherapy for resected pancreatic cancer: a multi-institutional retrospective analysis of 210 patients. Int J Radiat Oncol Biol Phys. 2010;77(3):734–42. doi: 10.1016/j.ijrobp.2009.09.010. [DOI] [PubMed] [Google Scholar]
  • 171.Valentini V, Morganti AG, Macchia G, et al. Intraoperative radiation therapy in resected pancreatic carcinoma: long-term analysis. Int J Radiat Oncol Biol Phys. 2008;70(4):1094–9. doi: 10.1016/j.ijrobp.2007.07.2346. [DOI] [PubMed] [Google Scholar]
  • 172.Kokubo M, Nishimura Y, Shibamoto Y, Sasai K, Kanamori S, Hosotani R, et al. Analysis of the clinical benefit of intraoperative radiotherapy in patients undergoing macroscopically curative resection for pancreatic cancer. International journal of radiation oncology, biology, physics. 2000;48(4):1081–7. doi: 10.1016/S0360-3016(00)00673-8. [DOI] [PubMed] [Google Scholar]
  • 173.Reni M, Panucci MG, Ferreri AJ, et al. Effect on local control and survival of electron beam intraoperative irradiation for resectable pancreatic adenocarcinoma. Int J Radiat Oncol Biol Phys. 2001;50(3):651–8. doi: 10.1016/S0360-3016(01)01470-5. [DOI] [PubMed] [Google Scholar]
  • 174.Cai S, Hong TS, Goldberg SI, et al. Updated long-term outcomes and prognostic factors for patients with unresectable locally advanced pancreatic cancer treated with intraoperative radiotherapy at the Massachusetts General Hospital, 1978 to 2010. Cancer. 2013;119(23):4196–204. doi: 10.1002/cncr.28329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Chen Y, Che X, Zhang J, et al. Long-term results of intraoperative electron beam radiation therapy for nonmetastatic locally advanced pancreatic cancer: retrospective cohort study, 7-year experience with 247 patients at the National Cancer Center in China. Medicine. 2016;95(38):e4861. doi: 10.1097/MD.0000000000004861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Jingu K, Tanabe T, Nemoto K, et al. Intraoperative radiotherapy for pancreatic cancer: 30-year experience in a single institution in Japan. Int J Radiat Oncol Biol Phys. 2012;83(4):e507–11. doi: 10.1016/j.ijrobp.2012.01.024. [DOI] [PubMed] [Google Scholar]
  • 177.Ma HB, Di ZL, Wang XJ, et al. Effect of intraoperative radiotherapy combined with external beam radiotherapy following internal drainage for advanced pancreatic carcinoma. World J Gastroenterol. 2004;10(11):1669–771. doi: 10.3748/wjg.v10.i11.1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Mohiuddin M, Regine WF, Stevens J, et al. Combined intraoperative radiation and perioperative chemotherapy for unresectable cancers of the pancreas. J Clin Oncol. 1995;13(11):2764–8. doi: 10.1200/JCO.1995.13.11.2764. [DOI] [PubMed] [Google Scholar]
  • 179.Nishimura Y, Hosotani R, Shibamoto Y, et al. External and intraoperative radiotherapy for resectable and unresectable pancreatic cancer: analysis of survival rates and complications. Int J Radiat Oncol, Biol, Phys. 1997;39(1):39–49. doi: 10.1016/S0360-3016(97)00295-2. [DOI] [PubMed] [Google Scholar]
  • 180.Willett CG, Del Castillo CF, Shih HA, et al. Long-term results of intraoperative electron beam irradiation (IOERT) for patients with unresectable pancreatic cancer. Ann Surg. 2005;241(2):295–9. doi: 10.1097/01.sla.0000152016.40331.bb. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Keane FK, Wo JY, Ferrone CR, et al. Intraoperative radiotherapy in the era of intensive neoadjuvant chemotherapy and chemoradiotherapy for pancreatic adenocarcinoma. Am J Clin Oncol. :9000. doi: 10.1097/COC.0000000000000336. [DOI] [PubMed] [Google Scholar]
  • 182.Conroy T, Bachet JB, Ayav A, et al. Current standards and new innovative approaches for treatment of pancreatic cancer. Eur J Cancer. 2016;57:10–22. doi: 10.1016/j.ejca.2015.12.026. [DOI] [PubMed] [Google Scholar]
  • 183.Hackert T, Sachsenmaier M, Hinz U, et al. Locally advanced pancreatic cancer: neoadjuvant therapy with folfirinox results in resectability in 60% of the patients. Ann Surg. 2016;264(3):457–63. doi: 10.1097/SLA.0000000000001850. [DOI] [PubMed] [Google Scholar]

Articles from ecancermedicalscience are provided here courtesy of ecancer Global Foundation

RESOURCES