Intraoperative Radiotherapy - CAM 80108

Description:
Intraoperative radiotherapy (IORT) is delivered directly to exposed tissues during surgery and may allow higher radiation doses by excluding nearby radiation dose-sensitive tissues. IORT can be delivered by electron beams produced by linear accelerators or high-dose rate brachytherapy.

For individuals who have rectal cancer who receive adjunctive IORT, the evidence includes a randomized controlled trial (RCT), nonrandomized comparative studies, and systematic reviews of these studies. Relevant outcomes are overall survival, disease-specific survival, change in disease status, and treatment-related morbidity. Adjunctive use of IORT as part of a multimodal treatment could permit an increase in radiation dose without increasing complications. However, a phase 3 RCT and meta-analysis of IORT for locally advanced rectal cancer did not find improved outcomes with IORT in combination with external-beam radiotherapy (EBRT) and surgery. Nonrandomized comparative studies and a meta- analysis of these studies have shown some benefit in health outcomes with adjunctive IORT for recurrent rectal cancer, but these studies are limited by a high risk of selection bias, heterogeneous patient populations, and heterogeneous delivery of other treatments. Large RCTs are needed to determine the effect of IORT with greater certainty.

For individuals who have gastric cancer who receive adjunctive IORT, the evidence includes RCTs and a systematic review of RCTs. Relevant outcomes are overall survival, disease-specific survival, change in disease status, and treatment-related morbidity. A meta-analysis of 8 RCTs found a benefit of IORT in locoregional control (but not overall survival) when used with EBRT. When IORT was administered without adjuvant EBRT in patients with stage III disease, overall survival improved. Thus, IORT might be considered an alternative to EBRT in patients undergoing surgery for stage III gastric cancer. Randomized studies comparing benefits and harms of the 2 treatments are needed to determine the efficacy of IORT with greater certainty. It cannot be determined whether IORT provides any benefit for overall survival in this patient population (gastric cancer patients) when used with EBRT.

For individuals who have soft tissue sarcomas who receive adjunctive IORT, the evidence includes a systematic review, a small RCT, and several nonrandomized comparative studies. Relevant outcomes are overall survival, disease-specific survival, change in disease status, and treatment-related morbidity. Overall, the study quality is low. The limited data suggest that IORT might improve local control and overall survival, but adverse events might outweigh any treatment benefit. RCTs are needed to determine the risks and benefits of IORT for soft tissue sarcomas with greater certainty.

For individuals who have gynecologic cancers who receive adjunctive IORT, the evidence includes a nonrandomized trial and case series. Relevant outcomes are overall survival, disease-specific survival, change in disease status, and treatment-related morbidity. The contribution of adjuvant IORT cannot be determined from the available literature.

For individuals who have head and neck cancers who receive adjunctive IORT, the evidence includes case series. Relevant outcomes are overall survival, disease-specific survival, change in disease status, and treatment-related morbidity. The strongest evidence is from a retrospective analysis of patients who had recurrent salivary gland carcinomas and were at risk of radiation toxicity due to prior treatment with EBRT. Some patients received IORT plus salvage surgery, and multivariate analysis found that use of IORT was a significant predictor of improved outcomes. Although these findings suggested an improvement in health outcomes for head and neck cancers that cannot be treated with EBRT due to toxicity, there was a high risk of selection bias in this study. Comparative trials are needed to determine the efficacy of IORT with greater certainty.

For individuals who have pancreatic cancer who receive adjunctive IORT, the evidence includes large case series, cohort studies, and systematic reviews of these studies. Relevant outcomes are overall survival, disease-specific survival, change in disease status, and treatment-related morbidity. The systematic reviews found no evidence that IORT was more effective than other therapies in treating pancreatic cancer. No evidence was identified that evaluated outcomes when IORT was and was not added to multimodal therapy.

For individuals who have renal cell carcinoma who receive adjunctive IORT, the evidence includes case series. Relevant outcomes are overall survival, disease-specific survival, change in disease status, and treatment-related morbidity. No controlled trials were identified to determine whether adjunctive IORT improves health outcomes when added to multimodal therapy with surgical resection and EBRT. Grade 3 or higher toxicity after IORT has been reported in a substantial percentage of patients.

For individuals who have glioblastoma or neuroblastoma or fibromatosis who receive adjunctive IORT, the evidence includes case series. Relevant outcomes are overall survival, disease-specific survival, change in disease status, and treatment-related morbidity. Compared with other therapies, it is unclear whether IORT improves overall survival. However, compared with historical controls, IORT for patients with previously untreated malignant gliomas had no survival benefit when given in conjunction with multimodal therapy. In addition, complication rates may be high. Comparative trials are needed to evaluate the safety and efficacy of this treatment modality.

Background 
Intraoperative radiotherapy (IORT) increases the intensity of radiation delivered directly to tumors. The tumor and associated tissues at risk for micrometastatic spread are directly visualized during surgery. IORT is delivered directly to the tumor, and normal or uninvolved tissues are not exposed to radiation because they are removed or shielded from the treatment field.

In the United States, certain racial/ethnic groups continue to be at an increased risk of developing or dying from particular cancers. Notably, Black men have the highest rate of new cancer diagnoses and Black men and women experience the highest rate of cancer-related death. Additionally, American Indians/Alaska Natives are disproportionally affected by kidney cancer and also have higher death rates from this cancer when compared to other racial/ethnic groups.

Regulatory Status 
The INTRABEAM® system was first approved for use by FDA for intracranial tumors in 1999 and was subsequently approved for whole body use in 2005. The INTRABEAM® spherical applicators are indicated for use with the INTRABEAM® system to deliver a prescribed dose of radiation to the treatment margin or tumor bed during intracavity or intraoperative radiotherapy treatments. The Mobetron® mobile election beam accelerator designed for use in the operating room received 510(k) marketing clearance in 1998. FDA product codes: JAD, LHN.

This policy does not address use of IORT for breast cancer; for that indication, see related Policy No. 80113.

Related Policies
80113  Accelerated Breast Irradiation After Breast-Conserving Surgery for Early Stage Breast Cancer and Breast Brachytherapy as Boost With Whole-Breast Irradiation
 

Policy:
Medically Necessary:
External beam intraoperative radiation therapy (IORT) is considered medically necessary as the sole source of boost therapy at the time of surgical excision for colorectal cancer, pancreatic cancer, pelvic cancers (for example, cervical or uterine),or soft tissue sarcomas, when either of the following criteria are met:

  • Tumor cannot be completely removed; or
  • Tumor has a high risk of recurring in surrounding tissues. 

Investigational and Not Medically Necessary: 
External beam intraoperative radiation therapy (IORT) is considered investigational and not medically necessary when the above criteria are not met, and for all other indications. 

Policy Guidelines
Effective in 2012, there are specific CPT codes for intraoperative radiotherapy: 

77424: Intraoperative radiation treatment delivery, X-ray, single treatment session
77425: Intraoperative radiation treatment delivery, electrons, single treatment session
77469: Intraoperative radiation treatment management. 

Prior to 2012, there was no CPT coding specific to radiotherapy given intraoperatively. The CPT codes for the radiotherapy (77261 – 77799) would have been used as appropriate. There was also a HCPCS code specific to intraoperative radiotherapy (S8049).

Benefit Application 
BlueCard®/National Account Issues
State or federal mandates (e.g., FEP) may dictate that all devices, drugs or biologics approved by the U.S. Food and Drug Administration (FDA) may not be considered investigational, and, thus, these devices may be assessed only on the basis of their medical necessity.

Rationale
Evidence reviews assess the clinical evidence to determine whether the use of a technology improves the net health outcome. Broadly defined, health outcomes are the length of life, quality of life, and ability to function-including benefits and harms. Every clinical condition has specific outcomes that are important to patients and managing the course of that condition. Validated outcome measures are necessary to ascertain whether a condition improves or worsens; and whether the magnitude of that change is clinically significant. The net health outcome is a balance of benefits and harms.

To assess whether the evidence is sufficient to draw conclusions about the net health outcome of a technology, 2 domains are examined: the relevance, and quality and credibility. To be relevant, studies must represent 1 or more intended clinical use of the technology in the intended population and compare an effective and appropriate alternative at a comparable intensity. For some conditions, the alternative will be supportive care or surveillance. The quality and credibility of the evidence depend on study design and conduct, minimizing bias and confounding that can generate incorrect findings. The randomized controlled trial (RCT) is preferred to assess efficacy; however, in some circumstances, nonrandomized studies may be adequate. Randomized controlled trials are rarely large enough or long enough to capture less common adverse events and long-term effects. Other types of studies can be used for these purposes and to assess generalizability to broader clinical populations and settings of clinical practice.

Promotion of greater diversity and inclusion in clinical research of historically marginalized groups (e.g., people of color [African-American, Asian, Black, Latino and Native American]; LGBTQIA (lesbian, gay, bisexual, transgender, queer, intersex, asexual); women; and people with disabilities [physical and invisible]) allows policy populations to be more reflective of and findings more applicable to our diverse members. While we also strive to use inclusive language related to these groups in our policies, use of gender-specific nouns (e.g., women, men, sisters, etc.) will continue when reflective of language used in publications describing study populations.

Intraoperative Radiotherapy for Rectal Cancer
Clinical Context and Therapy Purpose

The purpose of intraoperative radiotherapy (IORT) is to provide a treatment option that is an alternative to or an improvement on existing therapies for individuals with rectal cancer.

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with rectal cancer undergoing tumor resection.

Classification of surgical resection margins is listed in Table 1.

Table 1. General Surgical Resection Margin Classification  

Classification Definition
R0 Negative margins; no cancer cells detected in resected tissue
R1 Microscopic positive margin; cancer cells detected by microscope in resected tissue
R2 Macroscopic positive margin; tumor cells detected without microscope in resected tissue

Interventions
The therapy being considered is IORT. IORT delivers a fractional dose of radiation directly to the tumor/tumor bed while the area is exposed during surgery with the intent to minimize exposure to surrounding healthy tissues. Different IORT modalities are available that impact both the dose distribution and method of application. IORT techniques include electron beam IORT, high-dose-rate brachytherapy-based IORT, and low-energy X-ray IORT. Most clinical experience involves electron beam IORT.

IORT is performed with applicators and cones that attach to the treatment head of high-energy medical linear accelerators that are designed to direct radiation to defined surface structures. IORT can be used alone, but is more typically used in combination with other modalities such as surgical resection, EBRT, or chemotherapy.

Comparators
The following therapies and practices are currently being used for patients with rectal cancer: surgery alone, multimodal therapies (EBRT plus surgery or chemotherapy).

Most individuals receive preoperative or postoperative EBRT in addition to surgical resection of the tumor. Therefore, IORT would be considered an adjunctive treatment to multimodal treatment that includes surgery plus EBRT. For recurrent tumors already treated with EBRT, and tissue at risk for radiation toxicity (e.g., head and neck cancers), IORT is being evaluated in conjunction with surgery alone.

Outcomes
General outcomes of interest are overall survival (OS), disease-specific survival, and harms from treatment, specifically radiation toxicity (Table 2).

Table 2. Outcomes of Interest  

Outcomes Details Relevance
Overall survival Survival rate or proportion dead [Timing: 1 year to 10 years ] Considered the most reliable and preferred cancer endpoint
Disease-specific survival Disease/recurrence-free survival [Timing: 1 year to 10 years ] The most frequent use of this endpoint is in the adjuvant setting after definitive surgery or radiotherapy
Radiation toxicity Can be divided into acute, subacute, and chronic effects [Timing: Weeks (acute effects) or months (subacute, chronic) after treatment] Acute effects typically resolve within 2 weeks. Subacute and chronic effects include radiation pneumonitis, radiation-induced liver disease, fibrosis, and organ damage.

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Systematic Reviews
Primary, Advanced, and Recurrent Cancer

Four systematic reviews were identified that evaluated IORT for either primary locally advanced rectal cancer or locally recurrent colorectal or rectal cancer. Wiig et al. (2014) reviewed 18 studies on primary rectal cancer (including 1 RCT, 5 comparative trials, 7 trials without IORT) and 18 studies on locally recurrent rectal cancer (including 5 studies without IORT).1 Meta-analysis of the data was not performed due to heterogeneity in study designs and reporting. Mirnezami et al. (2013) included 29 studies (14 prospective, 15 retrospective) published between 1965 and 2011 (N = 3,003 ).2 Indications for IORT were locally advanced disease in 1,792 patients and locally recurrent disease in 1,211 patients with colorectal cancer. Liu et al. (2021) included 3 RCTs and 12 observational studies (N = 1,460) that evaluated IORT in both locally advanced and locally recurrent rectal cancer.3 Fahy et al. (2021) included 7 studies of patients with locally advanced and locally recurrent rectal cancer (N = 833).4

A comparison of the studies included in the systematic reviews is included in Table A-1. Characteristics and results of these reviews are summarized in Tables 3 and 4.

Table 3. Systematic Review Characteristics   

Study Dates Trials Participants N (Range) Design Duration
Wiig (2014)1 1990 to 2013 Primary cancer: 15; Recurrent cancer: 18 Patients with locally advanced rectal cancer (either primary or recurrent) Primary cancer: 4,272; Recurrent cancer: 1,174 (ranges not reported) RCTs (if available), comparative studies, non-comparative studies, non-IORT studies Up to 5 years
Mirnezami (2013)2 1991 to 2011 29 Patients with locally advanced colorectal cancer (either primary or recurrent) 3,003 (11 to 607) RCTs (if available), prospective and retrospective observational studies Up to 5 years
Liu (2021)3 1991 to 2020 15 Patients with rectal cancer 1,460 (ranges not reported) RCTs (if available), prospective and retrospective observational studies Up to 5 years
Fahy (2021)4 2000 to 2020 7 Patients with locally advanced and locally recurrent rectal cancer 833 (ranges not reported) RCTs (if available), prospective and retrospective observational studies Not reported

IORT: intraoperative radiotherapy; RCT: randomized controlled trial.

Table 4. Systematic Review Resultsa

Study OS DFS Local relapse
Wiig (2014)1 OS   5-year local control
Primary cancer      
Total N Not reported (20 studies)   Not reported (18 studies)
IORT, mean (range) 60 (28 to 76)   13 (2 to 35)
non-IORT, mean (range) 72 (52 to 85)   8 (5 to 9)
Locally recurrent cancer      
Total N NR (23 studies)   NR (12 studies)
IORT, mean (range) 25 (40 to 46)   49 (28 to 74)
non-IORT, mean (range) 19 (0 to 46)   81 (70 to 92)
Mirnezami (2013)2 5-year OS, IORT vs no IORT 5-year DFS, IORT vs no IORT 5-year local control, IORT vs no IORT
Total N 370 288 482
Pooled effect (95% CI) HR = 0.33 (0.2 to 0.54) HR = 0.51 (0.31 to 0.85) OR = 0.22 (0.05 to 0.86)
I(p) 0 (.001) 42% (.009) 68% (.03)
Range of N 19 to 167 37 to 167 19 to 167
Range of effect sizes 0.13 to 0.36 0.32 to 1.54 0.04 to 1.88
Liu (2021)3 5-year OS, IORT vs no IORT 5-year DFS, IORT vs no IORT 5-year local control, IORT vs no IORT
Total N NR (9 studies) NR (6 studies) NR (14 studies)
Pooled effect (95% CI) HR = 0.80 (0.60 to 1.06) HR = 0.94 (0.73 to 1.22) HR = 3.07 (1.66 to 5.66)
I(p) 0 (.740) 0 (.503) 70.9 (.000)
Range of effect sizes 0.31 to 2.31 0.81 to 1.93 0.74 to 17.53
Fahy (2021)4     Locoregional recurrence, IORT vs no IORT
Total N     833
Pooled effect (95% CI)     OR = 0.55 (0.27 to 1.14)
I(p)     55 (.11)
Range of N     19 to 99
Range of effect sizes     0.10 to 1.45

a Formal meta-analysis not conducted in Wiig (2014), instead mean (range) for outcomes were presented for the publications included. 
CI: confidence interval; DFS: disease-free survival; HR: hazard ratio; IORT: intraoperative radiotherapy; NR: not reported; OR: odds ratio; OS: overall survival.

Mirnezami et al. (2013) demonstrated significant survival and local control benefits with IORT in a mixed population of patients with locally advanced colorectal cancer (either primary or recurrent).2 More recently, however, Liu et al. (2021) did not demonstrate a 5-year OS or disease-free survival (DFS) benefit with IORT in patients with rectal cancer.3 IORT did, however, demonstrate benefit in 5-year local control. Fahy et al. (2021) also did not find a benefit with IORT for locoregional recurrence in a mixed population of patients with locally advanced and locally recurrent rectal cancer.4 Wiig et al. (2014) results suggested IORT provided no OS benefit for primary completely resected rectal cancers, with a possible reduction in local recurrence in cases of incomplete tumor resection.1 There was no evidence that IORT affected OS or local recurrence when used to treat locally recurrent rectal cancer.

Some analyses also reported outcomes for complications following IORT. Mirnezami et al. (2013) did not demonstrate an increased risk in total (odds ratio [OR] = 1.13; 95% confidence interval [CI], 0.77 to 1.65), urologic (OR = 1.35; 95% CI, 0.84 to 2.82), or anastomotic (OR = 0.94; 95% CI, 0.42 to 2.1) complications with IORT; however, increased wound complications were noted after IORT (OR = 1.86; 95% CI, 1.03 to 3.38; p = .049).2 Liu et al. (2021) did not find an increase in the risk of complications with IORT, including fistulae (OR = 0.79; 95% CI, 0.33 to 1.89), wound complication (OR = 1.21; 95% CI, 0.62 to 2.36), anastomotic leak (OR = 1.09; 95% CI, 0.59 to 2.02), or neurogenic bladder dysfunction (OR = 0.69; 95% CI, 0.31 to1.55).3 Likewise, Fahy et al. (2021) did not find an increased risk of complications with IORT, including wound infections (OR = 1.13; 95% CI, 0.50 to 2.54), pelvic abscess (OR = 1.01; 95% CI, 0.54 to 1.87), or anastomotic leak (OR = 1.60; 95% CI, 0.51 to 2.81).4 All reviews are limited by the risk of selection bias for IORT in nonrandomized studies, the variability in stages evaluated and IORT dosing, and high heterogeneity present for certain outcomes.

Randomized Controlled Trials
Locally Advanced Cancer

The available RCTs evaluating IORT for locally advanced rectal cancer are summarized in Table 5. No RCTs were identified that evaluated IORT for the management of locally recurrent rectal cancers.

Table 5. Summary of Key RCT Characteristics 

Study Countries Sites Dates Participants Interventions
          Active Comparator
Dubois (2011)5 France 7 1993 to 2001 142 patients with locally advanced rectal cancer (infiltrative rectal adenocarcinoma; T3 or T4 or N+, and M0) treated with preoperative radiotherapy IORT plus surgical resection (n = 73) Surgical resection alone (n = 69)
Masaki (2020)6 Japan 1 Not reported. Terminated in 2017 76 patients with locally advanced rectal cancer (M0) IORT plus resection of the rectum with total mesorectal excision (n = 38) Resection of the rectum with total mesorectal excision alone (n = 38)

IORT: intraoperative radiotherapy; RCT: randomzied controlled trial. 

Health outcome results for RCTs are summarized in Table 6. Additionally, in the Dubois et al. (2011) trial, postoperative complications were observed in 29.6% of patients in the IORT group and 19.1% of patients in the control group (p = .15).5 Specific, radiation-specific complications were not reported. In the Masaki et al. (2020) trial, the primary outcome of the study was to compare the pelvic sidewall recurrence rate between the groups.6, The trial was prematurely stopped in July 2017 because distant metastasis-free survivals were found to be significantly worse in the IORT group compared to the control group. Therefore, the authors concluded that IORT should not be recommended as a standard therapy to compensate for less radical resection for advanced lower rectal cancer.

Table 6. Summary of Key RCT Results

Study OS DFS Local relapse
Dubois (2011)5 Median Median Local control at 5 years (%)
N 140 140 140
IORT + surgical resection 88 months 80 months 91.8%
Surgical resection 106 months 89 months 92.8%
Difference Not reported (p = .2578) Not reported (p = .6037) Not reported (p = .6018)
Masaki (2020)6 5-year, 10-year, and 15-year OS 5-year, 10-year, and 15-year distant metastasis-free survival 5-year pelvic sidewall recurrence
N 76 76 76
IORT + surgical resection 71.5%, 61.7%, and 61.7% 57.5%, 53%, and 53% 12.4%
Surgical resection 81.8%, 73.8%, and 64.6% 76.8%, 76.8%, and 76.8% 8.3%
Difference (95% CI) OR = 1.264 (0.523 to 3.051); p = .603 OR = 2.554 (1.041 to 6.269); p = .041 OR = 1.350 (0.302 to 6.034); p = .694

CI: confidence interval; DFS: disease-free survival; IORT: intraoperative radiotherapy; OR: odds ratio; OS: overall survival; RCT: randomzied controlled trial.

The purpose of the limitations tables (see Tables 7 and 8) is to display notable gaps identified in each study. This information is synthesized as a summary of the body of evidence following each table and provides conclusions on the sufficiency of evidence supporting the position statement.

Table 7. Study Relevance Limitations

Study Populationa Interventionb Comparatorc Outcomesd Follow-Upe
Dubois (2011)5          
Masaki (2020)6 2. Staging of advanced rectal cancer not reported      

The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.
a Population key: 1. Intended use population unclear; 2. Study population is unclear; 3. Study population not representative of intended use; 4. Enrolled populations do not reflect relevant diversity; 5. Other.
b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest (e.g., proposed as an adjunct but not tested as such); 5: Other.
c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively; 5. Other.
d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. Incomplete reporting of harms; 4. Not establish and validated measurements; 5. Clinically significant difference not prespecified; 6. Clinically significant difference not supported; 7. Other.
e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms; 3. Other.

Table 8. Study Design and Conduct Limitations

Study Allocationa Blindingb Selective Reportingc Data Completenessd Powere Statisticalf
Dubois (2011)5   1. Patients and surgeons were not blinded to treatment assignment, though impractical for this study     3. Percent of local failures was smaller than expected, which may have reduced the power  
Masaki (2020)6   1. Patients and surgeons were not blinded to treatment assignment, though impractical for this study     3. Trial was terminated early likely reducing power

The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.
a Allocation key: 1. Participants not randomly allocated; 2. Allocation not concealed; 3. Allocation concealment unclear; 4. Inadequate control for selection bias; 5. Other.
b Blinding key: 1. Participants or study staff not blinded; 2. Outcome assessors not blinded; 3. Outcome assessed by treating physician; 4. Other.
c Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication; 4. Other.
d Data Completeness key: 1. High loss to follow-up or missing data; 2. Inadequate handling of missing data; 3. High number of crossovers; 4. Inadequate handling of crossovers; 5. Inappropriate exclusions; 6. Not intent to treat analysis (per protocol for noninferiority trials); 7. Other.
e Power key: 1. Power calculations not reported; 2. Power not calculated for primary outcome; 3. Power not based on clinically important difference; 4. Other.
f Statistical key: 1. Analysis is not appropriate for outcome type: (a) continuous; (b) binary; (c) time to event; 2. Analysis is not appropriate for multiple observations per patient; 3. Confidence intervals and/or p values not reported; 4. Comparative treatment effects not calculated; 5. Other.

Section Summary: Rectal Cancer
The evidence for IORT as part of a multimodal treatment approach in patients who have locally advanced (colo-)rectal cancer includes RCTs, nonrandomized comparative studies, and systematic reviews with meta-analyses of these studies. Adjunctive use of IORT could permit an increase in radiation dose without increasing complications. Available meta-analyses on IORT, in addition to standard therapy, for rectal cancer have combined together studies on both locally advanced primary and recurrent disease. Of the 2 systematic reviews that quantitatively pooled results, there was no benefit with the addition of IORT in terms of survival, but there was conflicting results on local control with one demonstrating an improvement in 5-year local control, while the other found no benefit in locoregional reoccurrence. In individuals with locally advanced primary rectal cancer only, 2 RCTs failed to show benefit with the addition of IORT in terms of local control or survival. For individuals with locally advanced primary or recurrent colorectal disease, one meta-analysis evaluating these populations together showed a significant benefit with the addition of IORT on local control, DFS, and OS. More data are needed to determine the effect of adjunctive IORT in each specific population of locally advanced disease (i.e., primary vs recurrent, rectal vs colorectal) with greater certainty.

Intraoperative Radiotherapy for Gastric Cancer
Clinical Context and Therapy Purpose

The purpose of IORT is to provide a treatment option that is an alternative to or an improvement on existing therapies for individuals with gastric cancer.

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with gastric cancer undergoing tumor resection.

Classification of surgical resection margins is listed in Table 9.

Table 9. General Surgical Resection Margin Classification

Classification Definition
R0 Negative margins; no cancer cells detected in resected tissue
R1 Microscopic positive margin; cancer cells detected by microscope in resected tissue
R2 Macroscopic positive margin; tumor cells detected without microscope in resected tissue

Interventions
The therapy being considered is IORT. IORT delivers a fractional dose of radiation directly to the tumor/tumor bed while the area is exposed during surgery with the intent to minimize exposure to surrounding healthy tissues. Different IORT modalities are available that impact both the dose distribution and method of application. IORT techniques include electron beam IORT, high-dose-rate brachytherapy-based IORT, and low-energy X-ray IORT. Most clinical experience involves electron beam IORT.

IORT is performed with applicators and cones that attach to the treatment head of high-energy medical linear accelerators that are designed to direct radiation to defined surface structures. IORT can be used alone, but is more typically used in combination with other modalities such as surgical resection, EBRT, or chemotherapy.

Comparators
The following therapies and practices are currently being used for patients with gastric cancer: surgery alone, multimodal therapies (EBRT plus surgery or chemotherapy).

Most patients receive preoperative or postoperative EBRT in addition to surgical resection of the tumor. Therefore, IORT would be considered an adjunctive treatment to multimodal treatment that includes surgery plus EBRT. For recurrent tumors already treated with EBRT, and tissue at risk for radiation toxicity (e.g., head and neck cancers), IORT is being evaluated in conjunction with surgery alone.

Outcomes
General outcomes of interest are OS, disease-specific survival, and harms from treatment, specifically radiation toxicity (Table 10).

Table 10. Outcomes of Interest

Outcomes Details Relevance
Overall survival Survival rate or proportion dead [Timing: 1 year to 10 years ] Considered the most reliable and preferred cancer endpoint
Disease-specific survival Disease/recurrence-free survival [Timing: 1 year to 10 years ] The most frequent use of this endpoint is in the adjuvant setting after definitive surgery or radiotherapy
Radiation toxicity Can be divided into acute, subacute, and chronic effects [Timing: Weeks (acute effects) or months (subacute, chronic) after treatment] Acute effects typically resolve within 2 weeks. Subacute and chronic effects include radiation pneumonitis, radiation-induced liver disease, fibrosis, and organ damage.

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Systematic Review

A meta-analysis by Yu et al. (2015) assessed 8 RCTs that used IORT for resectable gastric cancer.7 The literature search from 1990 through mid-2013 identified trials that assigned patients to surgery plus IORT or to surgery without IORT. Three studies also gave EBRT to both arms. Hazard ratios (HR) to describe the impact of adjuvant IORT on OS and locoregional control were obtained directly from the original studies or calculated from survival curves. Compiled data from 4 studies that reported OS revealed that IORT had no significant impact on OS (HR = 0.97; 95% CI, 0.75 to 1.26; p = .837). Notably, 3 of the 4 studies provided adjuvant EBRT. In another 3 studies that tested the efficacy of IORT for OS in patients with stage III disease, OS significantly improved (HR = 0.60; 95% CI, 0.40 to 0.89; p = .011). However, all 3 of these studies did not administer EBRT and used a higher dose of IORT than the other studies. The largest study in the meta-analysis included 292 patients with stage III disease. The HR for OS in this study was 0.54 (95% CI, 0.35 to 0.83). Significant improvement in locoregional control was observed in 4 studies that provided such data (HR = 0.40; 95% CI, 0.26 to 0.62; p < .001).

Section Summary: Gastric Cancer
A meta-analysis of 8 RCTs found a benefit of IORT in locoregional control but not OS when used in combination with EBRT. Three studies found improved OS in patients with stage III disease; however, none of the 3 studies provided EBRT. Randomized studies comparing the benefits and harms of IORT and EBRT are needed to determine the efficacy of IORT with greater certainty. It cannot be determined from the current literature whether IORT in patients with stage III disease provides any benefit for OS when used with EBRT.

Intraoperative Radiotherapy for Soft Tissue Sarcomas
Clinical Context and Therapy Purpose

The purpose of IORT is to provide a treatment option that is an alternative to or an improvement on existing therapies for individuals with soft tissue sarcoma.

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with soft tissue sarcoma undergoing tumor resection.

Classification of surgical resection margins is listed in Table 11.

Table 11. General Surgical Resection Margin Classification

Classification Definition
R0 Negative margins; no cancer cells detected in resected tissue
R1 Microscopic positive margin; cancer cells detected by microscope in resected tissue
R2 Macroscopic positive margin; tumor cells detected without microscope in resected tissue

Interventions
The therapy being considered is IORT. IORT delivers a fractional dose of radiation directly to the tumor/tumor bed while the area is exposed during surgery with the intent to minimize exposure to surrounding healthy tissues. Different IORT modalities are available that impact both the dose distribution and method of application. IORT techniques include electron beam IORT, high-dose-rate brachytherapy-based IORT, and low-energy x-ray IORT. Most clinical experience involves electron beam IORT.

IORT is performed with applicators and cones that attach to the treatment head of high-energy medical linear accelerators that are designed to direct radiation to defined surface structures. IORT can be used alone, but is more typically used in combination with other modalities such as surgical resection, EBRT, or chemotherapy.

Comparators
The following therapies and practices are currently being used for patients with soft tissue sarcoma: surgery alone, multimodal therapies (EBRT plus surgery or chemotherapy).

Most patients receive preoperative or postoperative EBRT in addition to surgical resection of the tumor. Therefore, IORT would be considered an adjunctive treatment to multimodal treatment that includes surgery plus EBRT. For recurrent tumors already treated with EBRT, and tissue at risk for radiation toxicity (eg, head and neck cancers), IORT is being evaluated in conjunction with surgery alone.

Outcomes
General outcomes of interest are OS, disease-specific survival, and harms from treatment, specifically radiation toxicity (Table 12).

Table 12. Outcomes of Interest

Outcomes Details Relevance
Overall survival Survival rate or proportion dead [Timing: 1 year to 10 years ] Considered the most reliable and preferred cancer endpoint
Disease-specific survival Disease/recurrence-free survival [Timing: 1 year to 10 years ] The most frequent use of this endpoint is in the adjuvant setting after definitive surgery or radiotherapy
Radiation toxicity Can be divided into acute, subacute, and chronic effects [Timing: Weeks (acute effects) or months (subacute, chronic) after treatment] Acute effects typically resolve within 2 weeks. Subacute and chronic effects include radiation pneumonitis, radiation-induced liver disease, fibrosis, and organ damage.

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Systematic Review

A systematic review by Skandarajah et al. (2009) highlights the potential value of IORT in the multimodal treatment of retroperitoneal sarcoma because these tumors are often close to dose-limiting structures, but reviewers noted that it is not without complications.8

Randomized Controlled Trial
One randomized trial (n = 35), reported by Sindelar et al. (1993), compared IORT plus low-dose (35- to 40-gray) postoperative EBRT with high-dose (50- to 55-gray) EBRT alone.9 The local recurrence rate was lower (40%) in the combined therapy group than in the EBRT-only group (80%), with no difference in OS. Patients who received IORT had fewer radiation enteritis events but had more disabling peripheral neuropathies.

Nonrandomized Comparative Studies
In a nonrandomized comparative study of 251 patients, 92 of whom received IORT, Lehnert et al. (2000) reported that IORT patients had more surgical complications and significantly more infectious complications; however, the IORT-treated patients had a 40% lower rate of local recurrence.10 IORT demonstrated effective tumor control in osteosarcoma.

A multicenter study by Calvo et al. (2014) compared outcomes from 159 patients who had soft tissue sarcomas of the extremity treated using IORT plus multimodal therapy with 95 patients treated using multimodal therapy without IORT.11 IORT was administered to patients who had close (< 1 cm) or positive surgical margins while patients with margins of 1 cm or greater were treated only with multimodal therapy. Use of IORT in the high-risk patients led to 5-year local control (82%) and OS rates (72%) that were similar to lower risk sarcoma patients treated without IORT. DFS (62%) remained modest due to the high risk of distant metastases. In multivariate analysis, only surgical margin resection was significantly associated with local control.

Stucky et al. (2014) reported on 63 consecutive patients with retroperitoneal sarcoma treated with surgery plus IORT (n = 37) or surgery-only (n = 26) between 1996 and 2011.12 Median follow-up was 45 months. The 5-year local control rate for patients receiving surgery plus IORT was 89% versus 46% for the surgery-only patients (p = .03). Survival did not differ as both groups had a 5-year OS rate of 60%. The contribution of IORT cannot be determined from this study.

Section Summary: Soft Tissue Sarcomas
The evidence on the use of adjunctive IORT for the treatment of soft tissue sarcomas includes a systematic review, a small RCT, and several nonrandomized comparative studies. Overall, study quality was low. The limited data available would suggest that IORT might improve local control and OS but adverse events might outweigh any treatment benefit. 

Intraoperative Radiotherapy for Gynecologic Cancers
Clinical Context and Therapy Purpose

The purpose of IORT is to provide a treatment option that is an alternative to or an improvement on existing therapies for individuals with gynecologic cancer.

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with gynecologic cancer undergoing tumor resection.

Classification of surgical resection margins is listed in Table 13.

Table 13. General Surgical Resection Margin Classification

Classification Definition
R0 Negative margins; no cancer cells detected in resected tissue
R1 Microscopic positive margin; cancer cells detected by microscope in resected tissue
R2 Macroscopic positive margin; tumor cells detected without microscope in resected tissue

Interventions
The therapy being considered is IORT. IORT delivers a fractional dose of radiation directly to the tumor/tumor bed while the area is exposed during surgery with the intent to minimize exposure to surrounding healthy tissues. Different IORT modalities are available that impact both the dose distribution and method of application. IORT techniques include electron beam IORT, high-dose-rate brachytherapy-based IORT, and low-energy X-ray IORT. Most clinical experience involves electron beam IORT.

IORT is performed with applicators and cones that attach to the treatment head of high-energy medical linear accelerators that are designed to direct radiation to defined surface structures. IORT can be used alone, but is more typically used in combination with other modalities such as surgical resection, EBRT, or chemotherapy.

Comparators
The following therapies and practices are currently being used for patients with gynecologic cancer: surgery alone, multimodal therapies (EBRT plus surgery or chemotherapy).

Most patients receive preoperative or postoperative EBRT in addition to surgical resection of the tumor. Therefore, IORT would be considered an adjunctive treatment to multimodal treatment that includes surgery plus EBRT. For recurrent tumors already treated with EBRT, and tissue at risk for radiation toxicity (e.g., head and neck cancers), IORT is being evaluated in conjunction with surgery alone.

Outcomes
General outcomes of interest are OS, disease-specific survival, and harms from treatment, specifically radiation toxicity (Table 14).

Table 14. Outcomes of Interest

Outcomes Details Relevance
Overall survival Survival rate or proportion dead [Timing: 1 year to 10 years ] Considered the most reliable and preferred cancer endpoint
Disease-specific survival Disease/recurrence-free survival [Timing: 1 year to 10 years ] The most frequent use of this endpoint is in the adjuvant setting after definitive surgery or radiotherapy
Radiation toxicity Can be divided into acute, subacute, and chronic effects [Timing: Weeks (acute effects) or months (subacute, chronic) after treatment] Acute effects typically resolve within 2 weeks. Subacute and chronic effects include radiation pneumonitis, radiation-induced liver disease, fibrosis, and organ damage.

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Observational Studies

In a phase 2 trial, Giorda et al. (2011) examined the use of radical surgery with IORT after chemotherapy in extracervical, locally advanced cancer patients.13 Between 2000 and 2007, 42 locally advanced cervical cancer patients were treated at a single center in Italy. EBRT was administered to the whole pelvic region in combination with chemotherapy. After EBRT and chemotherapy, 35 (83%) of 42 patients underwent radical surgery and IORT treatment. Five-year DFS and OS rates were 46% and 49%, respectively. DFS and OS were significantly longer when the residual tumor was absent or limited to the cervix. At follow-up, only 3 (9%) of 35 patients were alive and free of disease.

A case series of 67 patients with locally advanced (n = 31) and recurrent cervical cancer (n = 36) treated with IORT at a Spanish center was reported by Martinez-Monge et al. (2001).14 Previously unirradiated patients received preoperative chemoradiation. The 10-year control rate within the area treated with IORT was 69.4% for the entire group, 98.2% for the primary group, and 46.4% for the recurrent group. Control in the treated area correlated with margin status, amount of residual disease, and pelvic lymph node involvement. The overall incidence of toxic events attributable to IORT was 13.9%. The 10-year survival rate for the entire group was 34%, 58% for patients with primary disease, and 14% for those with recurrent disease. Patients, especially those with recurrent disease, with positive lymph nodes, parametrial involvement, and/or incomplete resection had poor local control, despite IORT at the doses used in the study.

Gao et al. (2011) evaluated clinical outcomes and toxicity of IORT plus EBRT in advanced and recurrent ovarian carcinoma.15 All 45 patients in this series underwent optimal cytoreductive surgery at a single center in China. At 5-year follow-up, local control was observed in 68.9%, with OS and DFS rates of 64% and 56%, respectively. The major complication was peripheral neuropathy, affecting 5 (11%) patients.

Chen et al. (2022) evaluated the feasibility and safety of IORT as an adjuvant therapy for recurrent gynecological cancer in a case series of 5 women at a single center in Taiwan (cervical cancer, n = 2; endometrial cancer, n = 2; uterine leiomyosarcoma, n = 1).16 Three women died during follow-up, 2 of which had local recurrence or progression of disease. The median recurrence-free survival was 13.8 months (95% CI, 1.6 to not estimable) and the median OS was 16.4 months (95% CI, 4.7 months to not estimable).

Section Summary: Gynecologic Cancer
The literature on IORT for gynecologic cancers consists of a nonrandomized trial and case series. The contribution of adjuvant IORT cannot be determined from these studies. OS rates in patients with locally advanced or recurrent disease are low and reported complications can be severe.

Intraoperative Radiotherapy for Head and Neck Cancers
Clinical Context and Therapy Purpose

The purpose of IORT is to provide a treatment option that is an alternative to or an improvement on existing therapies for individuals with head and neck cancer.

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with head and neck cancer undergoing tumor resection.

Classification of surgical resection margins is listed in Table 15.

Table 15. General Surgical Resection Margin Classification

Classification Definition
R0 Negative margins; no cancer cells detected in resected tissue
R1 Microscopic positive margin; cancer cells detected by microscope in resected tissue
R2 Macroscopic positive margin; tumor cells detected without microscope in resected tissue

Interventions
The therapy being considered is IORT. IORT delivers a fractional dose of radiation directly to the tumor/tumor bed while the area is exposed during surgery with the intent to minimize exposure to surrounding healthy tissues. Different IORT modalities are available that impact both the dose distribution and method of application. IORT techniques include electron beam IORT, high-dose-rate brachytherapy-based IORT, and low-energy x-ray IORT. Most clinical experience involves electron beam IORT.

IORT is performed with applicators and cones that attach to the treatment head of high-energy medical linear accelerators that are designed to direct radiation to defined surface structures. IORT can be used alone, but is more typically used in combination with other modalities such as surgical resection, EBRT, or chemotherapy.

Comparators
The following therapies and practices are currently being used for patients with head and neck cancer: surgery alone, multimodal therapies (EBRT plus surgery or chemotherapy).

Most patients receive preoperative or postoperative EBRT in addition to surgical resection of the tumor. Therefore, IORT would be considered an adjunctive treatment to multimodal treatment that includes surgery plus EBRT. For recurrent tumors already treated with EBRT, and tissue at risk for radiation toxicity (eg, head and neck cancers), IORT is being evaluated in conjunction with surgery alone.

Outcomes
General outcomes of interest are OS, disease-specific survival, and harms from treatment, specifically radiation toxicity (Table 16).

Table 16. Outcomes of Interest

Outcomes Details Relevance
Overall survival Survival rate or proportion dead [Timing: 1 year to 10 years ] Considered the most reliable and preferred cancer endpoint
Disease-specific survival Disease/recurrence-free survival [Timing: 1 year to 10 years ] The most frequent use of this endpoint is in the adjuvant setting after definitive surgery or radiotherapy
Radiation toxicity Can be divided into acute, subacute, and chronic effects [Timing: Weeks (acute effects) or months (subacute, chronic) after treatment] Acute effects typically resolve within 2 weeks. Subacute and chronic effects include radiation pneumonitis, radiation-induced liver disease, fibrosis, and organ damage.

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Observational Studies

Zeidan et al. (2011, 2012) reported on 2 case series of head and neck cancers. In the 2011 publication, they reported on the use of IORT for 231 patients with advanced cervical metastasis.17 OS rates at 1, 3, and 5 years were 58%, 34%, and 26%, respectively. Recurrence-free survival rates at 1, 3, and 5 years were 66%, 55%, and 49%, respectively. A second publication reviewed the use of IORT in 96 patients with primary or recurrent cancer of the parotid gland.18 Recurrence-free survival rates at 1, 3, and 5 years were 82%, 69%, and 65%, respectively. Rates of OS 1, 3, and 5 years after surgery and IORT were 88%, 66%, and 56%, respectively. Complications developed in 26 patients.

Thirty-four patients with recurrent head and neck cancer treated with IORT at another center were reported by Perry et al. (2010).19 At a median follow-up of 23 months (range, 6 to 54 months), 8 patients were alive and without evidence of disease. The 1- and 2-year estimates for in-field local progression-free survival (PFS) rates were 66% and 56%, respectively, with 13 (34%) in-field recurrences. Distant metastases-free survival rates were 81% and 62%, respectively, with 10 (29%) patients developing distant failure. Rates of OS at 1 and 2 years were 73% and 55%, respectively, with a median time to OS of 24 months.

Chen et al. (2008) reported on a retrospective study of 99 patients with locally recurrent salivary gland carcinomas treated surgically with or without IORT.20 All patients had previously been treated with surgery, and 82% had received postoperative EBRT. The median time from the initial surgery to local recurrence was 3.1 years. After salvage surgery, 37 (37%) patients received IORT. Reasons for IORT use were not clearly described in the report. For the entire patient population, the 1-, 3-, and 5-year estimates of local control were 88%, 75%, and 69%, respectively. Univariate analysis revealed predictors of local recurrence to be positive surgical margins, tumor size greater than 4 cm, and lack of IORT. Six of 37 patients treated with IORT experienced a local recurrence compared with 26 of 32 treated without IORT. At 5 years, the OS rate was 34%, and the DFS rate was 46%. The only predictor of DFS was the use of IORT, with a 5-year DFS rate of 61% in patients treated with IORT and 44% in patients without IORT. Complications were not analyzed.

A case series of 137 patients with persistent or recurrent salivary gland tumors treated with IORT after surgical resection was also reported by Chen et al. (2007).21 There is a potential for overlap of patients with the Chen et al. (2008) study described above. Eighty-three percent had previously received EBRT. Surgical margins were microscopically positive in 56 patients. Median follow-up among surviving patients was 41 months (range, 3 to 122 months). Estimates of in-field control after surgery and IORT at 1, 2, and 3 years were 70%, 64%, and 61%, respectively, and positive margins at the time of IORT predicted in-field failure. Three-year rates of locoregional control, distant metastasis-free survival, and OS were 51%, 46%, and 36%, respectively.

Section Summary: Head and Neck Cancers
The evidence on the use of IORT for head and neck cancers includes case series. The strongest evidence is from a retrospective study of patients who had recurrent salivary gland carcinomas and were at risk of radiation toxicity due to prior treatment with EBRT. In this study, multivariate analysis found that the use of IORT was a significant predictor of improved outcomes. However, the reasons for using or not using IORT were not clearly described, and there was a risk of selection bias.

Intraoperative Radiotherapy for Pancreatic Cancer
Clinical Context and Therapy Purpose

The purpose of IORT is to provide a treatment option that is an alternative to or an improvement on existing therapies for individuals with pancreatic cancer.

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with pancreatic cancer undergoing tumor resection.

Classification of surgical resection margins is listed in Table 17.

Table 17. General Surgical Resection Margin Classification

Classification Definition
R0 Negative margins; no cancer cells detected in resected tissue
R1 Microscopic positive margin; cancer cells detected by microscope in resected tissue
R2 Macroscopic positive margin; tumor cells detected without microscope in resected tissue

Interventions
The therapy being considered is IORT. IORT delivers a fractional dose of radiation directly to the tumor/tumor bed while the area is exposed during surgery with the intent to minimize exposure to surrounding healthy tissues. Different IORT modalities are available that impact both the dose distribution and method of application. IORT techniques include electron beam IORT, high-dose-rate brachytherapy-based IORT, and low-energy x-ray IORT. Most clinical experience involves electron beam IORT.

IORT is performed with applicators and cones that attach to the treatment head of high-energy medical linear accelerators that are designed to direct radiation to defined surface structures. IORT can be used alone, but is more typically used in combination with other modalities such as surgical resection, EBRT, or chemotherapy.

Comparators
The following therapies and practices are currently being used for patients with pancreatic cancer: surgery alone, multimodal therapies (EBRT plus surgery or chemotherapy).

Most patients receive preoperative or postoperative EBRT in addition to surgical resection of the tumor. Therefore, IORT would be considered an adjunctive treatment to multimodal treatment that includes surgery plus EBRT. For recurrent tumors already treated with EBRT, and tissue at risk for radiation toxicity (e.g., head and neck cancers), IORT is being evaluated in conjunction with surgery alone.

Outcomes
General outcomes of interest are OS, disease-specific survival, and harms from treatment, specifically radiation toxicity (Table 18).

Table 18. Outcomes of Interest

Outcomes Details Relevance
Overall survival Survival rate or proportion dead [Timing: 1 year to 10 years ] Considered the most reliable and preferred cancer endpoint
Disease-specific survival Disease/recurrence-free survival [Timing: 1 year to 10 years ] The most frequent use of this endpoint is in the adjuvant setting after definitive surgery or radiotherapy
Radiation toxicity Can be divided into acute, subacute, and chronic effects [Timing: Weeks (acute effects) or months (subacute, chronic) after treatment] Acute effects typically resolve within 2 weeks. Subacute and chronic effects include radiation pneumonitis, radiation-induced liver disease, fibrosis, and organ damage.

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Systematic Review

One recent systematic review by Jin et al. (2020) was identified that evaluated clinical outcomes in patients with resectable pancreatic cancer with or without IORT.22 The meta-analysis identified 15 pertinent articles for inclusion representing 401 patients undergoing pancreatic resection with IORT and 433 patients undergoing pancreatic resection only. Characteristics and results are summarized in Tables 19 and 20.

Table 19. Systematic Review Characteristics

Study Dates Trials Participants N (Range) Design Duration
Jin (2020)22 1990 – 2019 15 Patients with resectable pancreatic cancer (not metastatic or locally advanced) undergoing surgery with or without IORT 834 (11 to 203) Non-randomized controlled trials Not reported

IORT: intraoperative radiotherapy

Table 20. Systematic Review Results

Study OS Local relapse
Jin (2020)22    
Total N Not reported (13 studies) Not reported (8 studies)
Pooled effect (95% CI) MSR = 1.20 (1.06 to 1.37) RR = 0.70 (0.51 to 0.97)
I(p) 65.3% (.005) 36.8%(.03)
Range of N Not reported  
Range of effect sizes 0.57 to 3.54 0.14 to 0.96

CI: confidence interval; MSR: median survival rate; RR: relative risk; OS: overall survival.

Jin et al. (2020) found that patients receiving IORT had an improved median survival rate and a reduced risk of local recurrence compared to those who did not receive adjuvant IORT with moderate heterogeneity.22 The incidence of postoperative complications between the groups were not significantly different from each other (relative risk = 0.95; 95% CI, 0.73 to 1.23). Results of the meta-analysis were limited by the small sample sizes of the included studies, substantial heterogeneity, and the mostly retrospective design of the studies.

Case Series
Other larger retrospective evaluations of IORT in pancreatic cancer that evaluated patients with unresectable disease are summarized in Tables 21 and 22 below.

Table 21. Summary of Case Series Characteristics - Unresectable Disease

Study Country Participants Follow-Up, months
Chen (2016)23 China 247 patients with nonmetastatic locally advanced pancreatic cancer; the male to female ratio was 1.4 median, 10.1
Cai (2013)24 United States 194 patients (men, 53%; racial/ethnic composition not specified) with unresectable locally advanced pancreatic cancer median, 11.6
Harrison (2020)25 United States 158 patients (men, 56%; racial/ethnic composition not specified) with borderline resectable/locally advanced pancreatic cancer (132 patients receiving FOLIRINOX were evaluated for survival analysis NR
Sekigami (2021)26 United States 201 patients (men, 51%; White race, >90%) with borderline resectable/locally advanced pancreatic cancer who received total neoadjuvant therapy (FOLIRINOX with chemoradiation) and underwent resection between 2011 and 2019. Of the 201 patients evaluated, 88 received IORT following resection; of these, 69 underwent R0 and 19 underwent R1 resection. NR
Cho (2022)27 Korea 41 patients (men, 56%) with resectable pancreatic cancer median, 9

FOLIRINOX: folinic acid, fluorouracil, irinotecan, oxaliplatin; IORT: intraoperative radiotherapy; NR: not reported.

Table 22. Summary of Case Series Results - Unresectable Disease

Study Treatment OS PFS
Chen (2016)23 IORT delivered after palliative surgical procedures; postoperative adjuvant therapy (e.g., chemotherapy) was recommended for all patients Overall 1-, 2- and 3-year survival rates were 40%, 14%, and 7.2%. Median OS was 9 months. 1-, 2- and 3-year LPFS rates were 51.3%, 40.1%, and 34.6%. 1-, 2- and 3-year DMFS rates were 39.3%, 23.4%, and 11.9%.
Cai (2013)24 IORT as part of multimodal approach including pre-IORT EBRT and chemotherapy Overall 1-, 2- and 3-year survival rates were 49%, 16%, and 6%. Median OS was 12 months. 1-, 2- and 3-year LPFS rates were 61%, 41%, and 38%. 1-, 2- and 3-year DMFS rates were 49%, 28%, and 19%.
Harrison (2020)25 IORT as part of multimodal approach including neoadjuvant treatment prior to attempted resection with IORT Overall 1-, 2-, 4-year survival rates were 99%, 79%, and 47% for those receiving any form of resection plus IORT. Overall 1-, 2-, 4-year survival rates were 98%, 49%, 13% for those receiving IORT only. At time of study follow-up, 51% and 67% of patients had disease progression in the resection plus IORT and IORT only groups, respectively.
Sekigami (2021)26 IORT following total neoadjuvant therapy (FOLIRINOX with chemoradiation) and resection Among patients who received IORT, there was no difference in OS between patients who underwent R0 vs R1 resection: R0: 48 months, IQR 25 to not reached vs R1: 37 months, IQR 30 to 47; p = .307. Among patients who received IORT, there was no difference in DFS between patients who underwent R0 vs R1 resection: R0: 29 months, IQR 14 to 47 vs R1: 20 months, IQR 15 to 28; p = .114.
Cho (2022)27 IORT as part of multimodal approach including adjuvant gemcitabine-based chemotherapy 1 year OS: 94.1% The 1-year local control and distant control rates were 76.4% and 55.7%, respectively.

DFS: disease-free survival; DMFS: distant metastasis-free survival; EBRT: external beam radiotherapy; FOLIRINOX: folinic acid, fluorouracil, irinotecan, oxaliplatin; IORT: intraoperative radiotherapy; IQR: interquartile range; LPFS: local progression-free survival; NR: not reported; OS: overall survival; PFS: progression-free survival.

Section Summary: Pancreatic Cancer
The evidence on the use of IORT for pancreatic cancer includes large case series and a systematic review of non-randomized comparative studies. The systematic review found that in patients with resectable pancreatic cancer the addition of IORT to standard therapy was associated with improved median survival and reduced local recurrence; the evidence was limited by mostly smaller retrospective designs contributing to the review. However, the vast majority of patients present at diagnosis with more advanced disease, such as borderline resectable, locally advanced, or with distant metastases. One-year and 2-year OS rates of patients with unresectable pancreatic cancer ranged from 40% to 98% and 14% to 49%, respectively, in large case series. Lastly, 1 case series found IORT combined with surgical resection to be associated with increased survival compared to IORT alone in patients with positive or close margins, and another case series found that application of IORT following resection yields similar survival outcomes regardless of R0 (generally better prognosis) or R1 (generally worse prognosis) resection. 

Intraoperative Radiotherapy for Renal Cell Carcinoma
Clinical Context and Therapy Purpose

The purpose of IORT is to provide a treatment option that is an alternative to or an improvement on existing therapies for individuals with renal cell carcinoma (RCC).

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with RCC undergoing tumor resection.

Classification of surgical resection margins is listed in Table 23.

Table 23. General Surgical Resection Margin Classification

Classification Definition
R0 Negative margins; no cancer cells detected in resected tissue
R1 Microscopic positive margin; cancer cells detected by microscope in resected tissue
R2 Macroscopic positive margin; tumor cells detected without microscope in resected tissue

Interventions
The therapy being considered is IORT. IORT delivers a fractional dose of radiation directly to the tumor/tumor bed while the area is exposed during surgery with the intent to minimize exposure to surrounding healthy tissues. Different IORT modalities are available that impact both the dose distribution and method of application. IORT techniques include electron beam IORT, high-dose-rate brachytherapy-based IORT, and low-energy X-ray IORT. Most clinical experience involves electron beam IORT.

IORT is performed with applicators and cones that attach to the treatment head of high-energy medical linear accelerators that are designed to direct radiation to defined surface structures. IORT can be used alone, but is more typically used in combination with other modalities such as surgical resection, EBRT, or chemotherapy.

Comparators
The following therapies and practices are currently being used for patients with RCC: surgery alone, multimodal therapies (EBRT plus surgery or chemotherapy).

Most patients receive preoperative or postoperative EBRT in addition to surgical resection of the tumor. Therefore, IORT would be considered an adjunctive treatment to multimodal treatment that includes surgery plus EBRT. For recurrent tumors already treated with EBRT, and tissue at risk for radiation toxicity (e.g., head and neck cancers), IORT is being evaluated in conjunction with surgery alone.

Outcomes
General outcomes of interest are OS, disease-specific survival, and harms from treatment, specifically radiation toxicity (Table 24).

Table 24. Outcomes of Interest

Outcomes Details Relevance
Overall survival Survival rate or proportion dead [Timing: 1 year to 10 years ] Considered the most reliable and preferred cancer endpoint
Disease-specific survival Disease/recurrence-free survival [Timing: 1 year to 10 years ] The most frequent use of this endpoint is in the adjuvant setting after definitive surgery or radiotherapy
Radiation toxicity Can be divided into acute, subacute, and chronic effects [Timing: Weeks (acute effects) or months (subacute, chronic) after treatment] Acute effects typically resolve within 2 weeks. Subacute and chronic effects include radiation pneumonitis, radiation-induced liver disease, fibrosis, and organ damage.

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Observational Studies

Paly et al. (2014) reported on 98 advanced or locally recurrent RCC patients treated with IORT during nephrectomy at 9 different institutions during the period of 1985 to 2010.28 Pre- or postoperative EBRT was given to 62% of patients. Median follow-up time was 3.5 years for surviving patients. For advanced disease, the 5-year OS, disease-specific survival, and DFS rates were 37%, 41%, and 39%, respectively. For locally recurrent disease, the 5-year OS, disease-specific survival, and DFS rates were 55%, 60%, and 52%, and reported to be favorable to patients who had resection without IORT.

Calvo et al. (2013) reported on 20-year outcomes in 25 patients with advanced (n = 15) or recurrent (n = 10) RCC treated with IORT.29,Fifteen (60%) patients received perioperative EBRT. Surgical resection resulted in negative margins (R0) in 6 (24%) patients and residual microscopic disease (R1) in 19 (76%) patients. Median follow-up for surviving patients was 22.2 years (range, 3.6 to 26 ). OS and DFS rates at 5 and 10 years were 38% and 18% and 19% and 14%, respectively. Locoregional control (tumor bed or regional lymph nodes) and distant metastases-free survival rates at 5 years were 80% and 22%, respectively. Six (24%) patients experienced acute or late toxicities of grade 3 or higher using National Cancer Institute Common Toxicity Criteria version 4.

Hallemeier et al. (2012) reported on outcomes of a multimodality therapy combining maximal surgical resection, EBRT, and IORT for 22 patients with advanced or recurrent RCC.30 Surgical resection was R0 (negative margins) in 5 patients (23%) and R1 (residual microscopic disease) in 17 patients (77%). OS rates at 1, 5, and 10 years were 91%, 40%, and 35% and DFS rates at 1, 5, and 10 years were 64%, 31%, and 31%, respectively. Central recurrence (within the IORT field), locoregional relapse (tumor bed or regional lymph nodes), and distant metastases rates at 5 years were 9%, 27%, and 64%, respectively.

Section Summary: Renal Cell Carcinoma
The evidence on the use of IORT for RCC includes case series. No controlled trials were identified to determine whether adjunctive IORT improves health outcomes when added to multimodal therapy with surgical resection and EBRT. In a case series, grade 3 or higher toxicity was reported in 24% of patients after IORT.

Intraoperative Radiotherapy for Glioblastoma or Neuroblastoma or Fibromatosis
Clinical Context and Therapy Purpose

The purpose of IORT is to provide a treatment option that is an alternative to or an improvement on existing therapies for individuals with glioblastoma or neuroblastoma or fibromatosis.

The following PICO was used to select literature to inform this review.

Populations
The relevant population of interest is individuals with glioblastoma or neuroblastoma or fibromatosis undergoing tumor resection.

Classification of surgical resection margins is listed in Table 25.

Table 25. General Surgical Resection Margin Classification

Classification Definition
R0 Negative margins; no cancer cells detected in resected tissue
R1 Microscopic positive margin; cancer cells detected by microscope in resected tissue
R2 Macroscopic positive margin; tumor cells detected without microscope in resected tissue

Interventions
The therapy being considered is IORT. IORT delivers a fractional dose of radiation directly to the tumor/tumor bed while the area is exposed during surgery with the intent to minimize exposure to surrounding healthy tissues. Different IORT modalities are available that impact both the dose distribution and method of application. IORT techniques include electron beam IORT, high-dose-rate brachytherapy-based IORT, and low-energy x-ray IORT. Most clinical experience involves electron beam IORT.

IORT is performed with applicators and cones that attach to the treatment head of high-energy medical linear accelerators that are designed to direct radiation to defined surface structures. IORT can be used alone, but is more typically used in combination with other modalities such as surgical resection, EBRT, or chemotherapy.

Comparators
The following therapies and practices are currently being used for patients with glioblastoma or neuroblastoma or fibromatosis: surgery alone, multimodal therapies (EBRT plus surgery or chemotherapy).

Most patients receive preoperative or postoperative EBRT in addition to surgical resection of the tumor. Therefore, IORT would be considered an adjunctive treatment to multimodal treatment that includes surgery plus EBRT. For recurrent tumors already treated with EBRT, and tissue at risk for radiation toxicity (e.g., head and neck cancers), IORT is being evaluated in conjunction with surgery alone.

Outcomes
General outcomes of interest are OS, disease-specific survival, and harms from treatment, specifically radiation toxicity (Table 26).

Table 26. Outcomes of Interest

Outcomes Details Relevance
Overall survival Survival rate or proportion dead [Timing: 1 year to 10 years ] Considered the most reliable and preferred cancer endpoint
Disease-specific survival Disease/recurrence-free survival [Timing: 1 year to 10 years ] The most frequent use of this endpoint is in the adjuvant setting after definitive surgery or radiotherapy
Radiation toxicity Can be divided into acute, subacute, and chronic effects [Timing: Weeks (acute effects) or months (subacute, chronic) after treatment] Acute effects typically resolve within 2 weeks. Subacute and chronic effects include radiation pneumonitis, radiation-induced liver disease, fibrosis, and organ damage.

Study Selection Criteria
Methodologically credible studies were selected using the following principles:

  • To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs.
  • In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.
  • To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.
  • Studies with duplicative or overlapping populations were excluded.

Review of Evidence
Glioblastoma
Observational Studies

Nemoto et al. (2002) reported on treatment with IORT for 32 patients with previously untreated malignant gliomas over a 10-year period.31 Patients also had postoperative radiotherapy. Eleven patients had histologic diagnoses of anaplastic astrocytoma, and 21 had glioblastoma. Median survival time was 24.7 months in the anaplastic astrocytoma group and 33.6 months for matched historical controls. Differences in 1-, 2-, and 5-year survival rates between IORT-treated patients and historical controls were also not statistically significant. In the glioblastoma group, median survival was 13.3 months for IORT-treated patients and 14.6 months for matched controls. Data on 1-, 2-, and 5-year survival rates also did not differ significantly between groups.

Sarria et al. (2020) reported on an international, retrospective, pooled analysis of patients with suspected glioblastoma/high-grade glioma treated with low-energy IORT, in addition to standard of care, across 5 institutions in 3 countries (Germany, Peru, and China).32 All patients received standard of care therapy and adjuvant therapies that included EBRT and temozolomide chemotherapy. A total of 51 patients were evaluated and followed for a median of 18 months. The 1-, 2-, and 3-year OS rates were 79.5%, 38.7% and 25.6% respectively (median survival time, 18 months). The 1-, 2-, and 3-year PFS rates were 46.2%, 29.4%, and 5.9%, respectively (median PFS, 11.4 months). The median local PFS was 16 months. Radionecrosis was observed in 13 patients (25.5%).

Section Summary: Glioblastoma
Compared with historical controls, IORT for patients with previously untreated malignant gliomas had no survival benefit when given as an adjunct to surgery and EBRT. An international retrospective pooled analysis of patients treated with IORT in addition to standard of care reported 1- and 2-year OS rates of 79.5% and 38.7%.

Neuroblastoma
Observational Study

Rich et al. (2011) reported on their experience using IORT after re-resection in patients with locally recurrent or persistent high-risk neuroblastomas.33 They retrospectively reviewed 44 consecutive patients who received IORT at a single institution between 2000 and 2009 after gross total resection of the recurrent or persistent tumor. Median follow-up after IORT was 10.5 months. Each patient had received prior chemotherapy and surgery, and 94.5% had received EBRT. Median OS was 18.7 months (95% CI, 11.7 to 25.6), with a 50.4% probability of local control.

Section Summary: Neuroblastoma
No controlled trials were identified. There is insufficient evidence to evaluate the efficacy of IORT as an adjunct to multimodal therapy for neuroblastomas.

Fibromatosis
Observational Study

Roeder et al. (2010) reviewed the outcomes of 30 patients (31 lesions) with aggressive fibromatosis who were treated with IORT after surgery.34 Treatment with IORT was undertaken to avoid mutilating surgical procedures when complete surgical removal seemed to be unlikely or impossible. The median age was 31 years (range, 13 to 59 ). Resection status was a close margin in 6 lesions, microscopically positive in 13, and macroscopically positive in 12. The median tumor size was 9 cm. Twenty-five (83%) patients received additional EBRT. After a median follow-up of 32 months (range, 3 to 139), no disease-related deaths occurred. Five local recurrences were reported, resulting in actutimes 3-year local control rates of 82% overall and 91% inside the IORT areas. Trends toward improved local control were seen for age (> 31 years) and negative surgical margins, but none of these factors were statistically significant. Perioperative complications were found in 6 patients (wound healing disturbances in 5 patients, venous thrombosis in 1 patient). Late toxicity was seen in 5 (17%) patients.

Section Summary: Fibromatosis
Although the local control rate for aggressive fibromatosis is high in patients who have had incomplete surgery and EBRT, no controlled trials were identified that evaluated whether IORT improves survival. Late toxicity was observed with the combined treatment in 17% of patients.

The purpose of the following information is to provide reference material. Inclusion does not imply endorsement or alignment with the evidence review conclusions.

Clinical Input From Physician Specialty Societies and Academic Medical Centers
While the various physician specialty societies and academic medical centers may collaborate with and make recommendations during this process, through the provision of appropriate reviewers, input received does not represent an endorsement or position statement by the physician specialty societies or academic medical centers, unless otherwise noted.

2009 Input
In response to requests, input was received from 1 physician specialty society and 2 academic medical centers (6 reviewers) while this policy was under review in 2009. Input was quite variable, with some supporting use of intraoperative radiotherapy for multiple indications and others considering it investigational. The strongest support was for rectal cancer.

Practice Guidelines and Position Statements
Guidelines or position statements will be considered for inclusion in Supplemental Information if they were issued by, or jointly by, a U.S. professional society, an international society with U.S. representation, or National Institute for Health and Care Excellence (NICE). Priority will be given to guidelines that are informed by a systematic review, include strength of evidence ratings, and include a description of management of conflict of interest.

American Brachytherapy Society
In 2019, the American Brachytherapy Society consensus statement on intraoperative radiotherapy (IORT) provides recommendations for patient selection for IORT.35, Table 27 summarizes their recommendations based on cancer type. The consensus statement did not rate evidence or strength of recommendations.

Table 27. Consensus statement on Use of IORT

Cancer site Recommendation
Breast cancer Monotherapy should not be offered unless in the context of a prospective clinical trial. Use as a boost technique can be considered in patients requiring a tumor bed boost.
CNS, brain metastases Can be considered for selected patients
CNS, high-grade gliomas Can be considered for selected patients
Colorectal Consider in cases with concern for positive margins. "IORT can be considered at the time of surgical resection of locally advanced or recurrent colorectal cancer in cases with concern for a positive margin, particularly when pelvic EBRT has already been delivered. A dose of 15 Gy in a single treatment to 5 mm depth in tissue using IORT-HDR has been used"
Gynecologic Consider in recurrent cases with concerns for close/positive margins. "IORT can be considered at the time of surgical resection for isolated recurrent gynecologic cancer in cases with concern for residual microscopic disease. IORT after chemoradiation and surgery for primary management of locally advanced cervical cancer should not be used off protocol."
Head and neck Can consider in selected patients
Pancreas Consider in cases with concerns for close/positive margins
Pediatric cancers Consider for pediatric sarcomas upfront if concern for close/positive margins or in recurrent sarcomas
Sarcoma, extremity Consider in situations with close/positive margins or recurrence with reirradiation
Sarcoma, retroperitoneal Consider in conjunction with preoperative EBRT, especially if close/positive margins are expected
Thorax Can be considered in selected patients. "IORT can be considered at the time of surgical resection in cases with concern for a positive margin. Intraoperative LDR brachytherapy may improve local control outcomes in patients undergoing sublobar resections for stage I NSCLC when there is a concern for a positive margin."

CNS: central nervous system; EBRT: external beam radiation therapy; Gy: gray; HDR: high dose radiation; IORT: intraoperative radiation therapy; LDR: low dose radiation; NSCLC: non-small cell lung cancer.

National Comprehensive Cancer Network
Table 28 lists the National Comprehensive Cancer Network guidelines on the use of IORT for the treatment of various cancers relevant to this evidence review.

Table 28. Recommendations for the Use of IORT

Cancer Site Version Recommendation COR
Central Nervous System v.1.202336 IORT is not addressed for the management of glioblastoma. NA
Cervical v.1.202337 IORT "is particularly useful in patients with recurrent disease within a previously radiated volume. During IORT, overlying normal tissue (such as bowel or other viscera) can be manually displaced from the region at risk." 3
Colon v.2.2023 38 IORT "if available, may be considered for patients with T4 or recurrent cancers as an additional boost." 2A
Gastric v.1.2023 39 IORT is not addressed. NA
Head/neck v.2.2023 40 "In certain rare circumstances, reirradiation with IORT or brachytherapy may be considered in high-volume centers with expertise in these techniques." 2A
Ovarian v.1.2023 41 IORT is not addressed. NA
Pancreatic v.1.2023 42 "Overall, there is no clear established role for IORT in patients with pancreatic cancer, and the panel believes it should only be performed at specialized centers." NA
Rectal v.3.2023 43 IORT "if available, may be considered for very close or positive margins after resection, as an additional boost, especially for patients with T4 or recurrent cancers." 2A
Renal v.4.2023 44 IORT is not addressed. NA
Soft tissue sarcoma v.2.2023 45 For patients with resectable disease, consider boost with IORT for known or suspected positive margins at the time of surgery "10 – 12.5 Gy for microscopic positive disease" and "15 Gy for gross disease". 2A
Uterine v.2.202346 Treatment of recurrent or metastatic disease:

"For patients previously treated with brachytherapy only at the recurrence site, surgery with (or without) IORT is recommended."

"For patients previously treated with EBRT at the recurrence site, recommended therapy for isolated relapse includes: 1) surgery with (or without) IORT plus or minus systemic therapy."

For local recurrence in the vaginal/pelvis that is negative for distant metastatic disease:

"Surgical and RT treatment pathways are provided. The surgical pathway for treating local recurrence in patients without prior RT exposure includes the option for IORT."

"Patients with local recurrence who have had prior RT exposure can be treated with 1) surgery with the option of IORT with (or without) systemic therapy; 2) systemic therapy; or 3) selected reirradiation with EBRT and/or brachytherapy."
3 for IORT

COR: category of recommendation; EBRT: external beam radiation therapy; Gy: gray; IORT: intraoperative radiotherapy; NA: not applicable; RT: radiotherapy.

U.S. Preventive Services Task Force Recommendations
Not applicable

Ongoing and Unpublished Clinical Trials
Some currently unpublished trials that might influence this review are listed in Table 29.

Table 29. Summary of Key Trials

NCT No. Trial Name Planned Enrollment Completion Date
Ongoing      
NCT05181488 A Prospective, Phase II Study Evaluating the Efficacy of Intraoperative Radiotherapy After Neoadjuvant Chemotherapy in Patients With Resectable Pancreatic Cancer 80 Apr 2026
NCT02685605 A Multicenter Randomized Phase III Trial on INTraoperative RAdiotherapy in Newly Diagnosed GliOblastoma Multiforme (INTRAGO II) 314 Mar 2024
NCT04681677 Phase II Study of Patients With Recurrent Glioblastoma Multiforme Treated With Maximal Safe Neurosurgical Resection and Intra-Operative Radiation Therapy (IORT) Using the Xoft Axxent Electronic Brachytherapy System and Bevacizumab 100 Feb 2027
NCT04847284 Intraoperative Radiotherapy in Patients With Brain Metastases 25 Mar 2024

GBM: glioblastoma; IORT: intraoperative radiotherapy; NCT: national clinical trial.

References 

  1. Wiig JN, Giercksky KE, Tveit KM. Intraoperative radiotherapy for locally advanced or locally recurrent rectal cancer: Does it work at all?. Acta Oncol. Jul 2014; 53(7): 865-76. PMID 24678823
  2. 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. Mar 2013; 22(1): 22-35. PMID 23270946
  3. Liu B, Ge L, Wang J, et al. Efficacy and safety of intraoperative radiotherapy in rectal cancer: A systematic review and meta-analysis. World J Gastrointest Oncol. Jan 15 2021; 13(1): 69-86. PMID 33510850
  4. Fahy MR, Kelly ME, Power Foley M, et al. The role of intraoperative radiotherapy in advanced rectal cancer: a meta-analysis. Colorectal Dis. Aug 2021; 23(8): 1998-2006. PMID 33905599
  5. Dubois JB, Bussieres E, Richaud P, et al. Intra-operative radiotherapy of rectal cancer: results of the French multi-institutional randomized study. Radiother Oncol. Mar 2011; 98(3): 298-303. PMID 21339010
  6. Masaki T, Matsuoka H, Kishiki T, et al. Intraoperative radiotherapy for resectable advanced lower rectal cancer-final results of a randomized controlled trial (UMIN000021353). Langenbecks Arch Surg. May 2020; 405(3): 247-254. PMID 32347365
  7. Yu WW, Guo YM, Zhang Q, et al. Benefits from adjuvant intraoperative radiotherapy treatment for gastric cancer: A meta-analysis. Mol Clin Oncol. Jan 2015; 3(1): 185-189. PMID 25469292
  8. Skandarajah AR, Lynch AC, Mackay JR, et al. The role of intraoperative radiotherapy in solid tumors. Ann Surg Oncol. Mar 2009; 16(3): 735-44. PMID 19142683
  9. Sindelar WF, Kinsella TJ, Chen PW, et al. Intraoperative radiotherapy in retroperitoneal sarcomas. Final results of a prospective, randomized, clinical trial. Arch Surg. Apr 1993; 128(4): 402-10. PMID 8457152
  10. Lehnert T, Schwarzbach M, Willeke F, et al. Intraoperative radiotherapy for primary and locally recurrent soft tissue sarcoma: morbidity and long-term prognosis. Eur J Surg Oncol. Nov 2000; 26 Suppl A: S21-4. PMID 11130875
  11. 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. Oct 2014; 190(10): 891-8. PMID 24715241
  12. 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. Jun 2014; 109(8): 798-803. PMID 24862926
  13. 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. May 2011; 37(5): 442-7. PMID 21492777
  14. Martínez-Monge R, Jurado M, Aristu JJ, et al. Intraoperative electron beam radiotherapy during radical surgery for locally advanced and recurrent cervical cancer. Gynecol Oncol. Sep 2001; 82(3): 538-43. PMID 11520152
  15. Gao Y, Liu Z, Chen X, et al. Intraoperative radiotherapy electron boost in advanced and recurrent epithelial ovarian carcinoma: a retrospective study. BMC Cancer. Oct 11 2011; 11: 439. PMID 21989202
  16. Chen HH, Hou PY, Ting WH, et al. Feasibility and Safety of Intraoperative Radiotherapy with Low Energy X-ray Photon Therapy for Recurrent Gynecological Cancer: A Case Series. Life (Basel). May 05 2022; 12(5). PMID 35629353
  17. Zeidan YH, Yeh A, Weed D, et al. Intraoperative radiation therapy for advanced cervical metastasis: a single institution experience. Radiat Oncol. Jun 15 2011; 6: 72. PMID 21676211
  18. Zeidan YH, Shiue K, Weed D, et al. Intraoperative radiotherapy for parotid cancer: a single-institution experience. Int J Radiat Oncol Biol Phys. Apr 01 2012; 82(5): 1831-6. PMID 21514074
  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. Mar 15 2010; 76(4): 1140-6. PMID 19560882
  20. Chen AM, Garcia J, Bucci MK, et al. Recurrent salivary gland carcinomas treated by surgery with or without intraoperative radiation therapy. Head Neck. Jan 2008; 30(1): 2-9. PMID 17828788
  21. 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. Jan 01 2007; 67(1): 122-9. PMID 17084543
  22. Jin L, Shi N, Ruan S, et al. The role of intraoperative radiation therapy in resectable pancreatic cancer: a systematic review and meta-analysis. Radiat Oncol. Apr 09 2020; 15(1): 76. PMID 32272945
  23. 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 (Baltimore). Sep 2016; 95(38): e4861. PMID 27661028
  24. 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. Dec 01 2013; 119(23): 4196-204. PMID 24006012
  25. Harrison JM, Wo JY, Ferrone CR, et al. Intraoperative Radiation Therapy (IORT) for Borderline Resectable and Locally Advanced Pancreatic Ductal Adenocarcinoma (BR/LA PDAC) in the Era of Modern Neoadjuvant Treatment: Short-Term and Long-Term Outcomes. Ann Surg Oncol. May 2020; 27(5): 1400-1406. PMID 31758284
  26. Sekigami Y, Michelakos T, Fernandez-Del Castillo C, et al. Intraoperative Radiation Mitigates the Effect of Microscopically Positive Tumor Margins on Survival Among Pancreatic Adenocarcinoma Patients Treated with Neoadjuvant FOLFIRINOX and Chemoradiation. Ann Surg Oncol. Aug 2021; 28(8): 4592-4601. PMID 33393047
  27. Cho Y, Kim JW, Kim HS, et al. Intraoperative Radiotherapy for Resectable Pancreatic Cancer Using a Low-Energy X-Ray Source: Postoperative Complications and Early Outcomes. Yonsei Med J. May 2022; 63(5): 405-412. PMID 35512742
  28. Paly JJ, Hallemeier CL, Biggs PJ, et al. Outcomes in a multi-institutional cohort of patients treated with intraoperative radiation therapy for advanced or recurrent renal cell carcinoma. Int J Radiat Oncol Biol Phys. Mar 01 2014; 88(3): 618-23. PMID 24411190
  29. Calvo FA, Sole CV, Martinez-Monge R, et al. Intraoperative EBRT and resection for renal cell carcinoma : twenty-year outcomes. Strahlenther Onkol. Feb 2013; 189(2): 129-36. PMID 23223810
  30. 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. Apr 01 2012; 82(5): 1938-43. PMID 21514065
  31. Nemoto K, Ogawa Y, Matsushita H, et al. Intraoperative radiation therapy (IORT) for previously untreated malignant gliomas. BMC Cancer. 2002; 2: 1. PMID 11818027
  32. Sarria GR, Sperk E, Han X, et al. Intraoperative radiotherapy for glioblastoma: an international pooled analysis. Radiother Oncol. Jan 2020; 142: 162-167. PMID 31629553
  33. Rich BS, McEvoy MP, LaQuaglia MP, et al. Local control, survival, and operative morbidity and mortality after re-resection, and intraoperative radiation therapy for recurrent or persistent primary high-risk neuroblastoma. J Pediatr Surg. Jan 2011; 46(1): 97-102. PMID 21238648
  34. Roeder F, Timke C, Oertel S, et al. Intraoperative electron radiotherapy for the management of aggressive fibromatosis. Int J Radiat Oncol Biol Phys. Mar 15 2010; 76(4): 1154-60. PMID 19647952
  35. Tom MC, Joshi N, Vicini F, et al. The American Brachytherapy Society consensus statement on intraoperative radiation therapy. Brachytherapy. 2019; 18(3): 242-257. PMID 31084904
  36. National Comprehensive Cancer Network. Central nervous system cancers. Version 1.2023. Updated March 24, 2023. https://www.nccn.org/professionals/physician_gls/pdf/cns.pdf. Accessed May 22, 2023.
  37. National Comprehensive Cancer Network. Cervical cancer. Version 1.2023. Updated April 28, 2023. https://www.nccn.org/professionals/physician_gls/pdf/cervical.pdf. Accessed June 1, 2023.
  38. National Comprehensive Cancer Network. Colon cancer. Version 2.2023. Updated April 25, 2023. https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf. Accessed May 31, 2023.
  39. National Comprehensive Cancer Network. Gastric cancer. Version 1.2023. Updated March 10, 2023. https://www.nccn.org/professionals/physician_gls/pdf/gastric.pdf. Accessed May 30, 2023.
  40. National Comprehensive Cancer Network. Head and neck cancers. Version 2.2023. Updated May 15, 2023. https://www.nccn.org/professionals/physician_gls/pdf/head-and-neck.pdf. Accessed May 29, 2023.
  41. National Comprehensive Cancer Network. Ovarian cancer. Version 1.2023. Updated February 22, 2023. https://www.nccn.org/professionals/physician_gls/pdf/ovarian.pdf. Accessed May 28, 2023.
  42. National Comprehensive Cancer Network. Pancreatic adenocarcinoma. Version 1.2023. Updated May 4, 2023. https://www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf. Accessed May 27, 2023.
  43. National Comprehensive Cancer Network. Rectal cancer. Version 3.2023. Updated May 26, 2023. https://www.nccn.org/professionals/physician_gls/pdf/rectal.pdf. Accessed May 26, 2023.
  44. National Comprehensive Cancer Network. Kidney cancer. Version 4.2023. Updated January 18, 2023. https://www.nccn.org/professionals/physician_gls/pdf/kidney.pdf. Accessed May 25, 2023.
  45. National Comprehensive Cancer Network. Soft tissue sarcoma. Version 2.2023. Updated April 25, 2023. https://www.nccn.org/professionals/physician_gls/pdf/sarcoma.pdf. Accessed May 24, 2023.
  46. National Comprehensive Cancer Network. Uterine neoplasms. Version 2. 2023. Updated April 28, 2023. https://www.nccn.org/professionals/physician_gls/pdf/uterine.pdf. Accessed May 23, 2023.
  47. Alberda WJ, Verhoef C, Nuyttens JJ, et al. Intraoperative radiation therapy reduces local recurrence rates in patients with microscopically involved circumferential resection margins after resection of locally advanced rectal cancer. Int J Radiat Oncol Biol Phys. Apr 01 2014; 88(5): 1032-40. PMID 24661656
  48. Bosset JF, Collette L, Calais G, et al. Chemotherapy with preoperative radiotherapy in rectal cancer. N Engl J Med. Sep 14 2006; 355(11): 1114-23. PMID 16971718
  49. Díaz-González JA, Calvo FA, Cortés J, et al. Prognostic factors for disease-free survival in patients with T3-4 or N+ rectal cancer treated with preoperative chemoradiation therapy, surgery, and intraoperative irradiation. Int J Radiat Oncol Biol Phys. Mar 15 2006; 64(4): 1122-8. PMID 16406393
  50. Ferenschild FT, Vermaas M, Nuyttens JJ, et al. Value of intraoperative radiotherapy in locally advanced rectal cancer. Dis Colon Rectum. Sep 2006; 49(9): 1257-65. PMID 16912909
  51. Gérard JP, Conroy T, Bonnetain F, et al. Preoperative radiotherapy with or without concurrent fluorouracil and leucovorin in T3-4 rectal cancers: results of FFCD 9203. J Clin Oncol. Oct 01 2006; 24(28): 4620-5. PMID 17008704
  52. Harris GJ, Senagore AJ, Lavery IC, et al. Factors affecting survival after palliative resection of colorectal carcinoma. Colorectal Dis. Jan 2002; 4(1): 31-35. PMID 12780652
  53. 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. Jul 1996; 39(7): 774-9. PMID 8674370
  54. Krempien R, Roeder F, Oertel S, et al. Long-term results of intraoperative presacral electron boost radiotherapy (IOERT) in combination with total mesorectal excision (TME) and chemoradiation in patients with locally advanced rectal cancer. Int J Radiat Oncol Biol Phys. Nov 15 2006; 66(4): 1143-51. PMID 16979835
  55. 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. Jun 2010; 21(6): 1279-1284. PMID 19889621
  56. Kusters M, Holman FA, Martijn H, et al. Patterns of local recurrence in locally advanced rectal cancer after intra-operative radiotherapy containing multimodality treatment. Radiother Oncol. Aug 2009; 92(2): 221-5. PMID 19339070
  57. Klink CD, Binnebösel M, Holy R, et al. Influence of intraoperative radiotherapy (IORT) on perioperative outcome after surgical resection of rectal cancer. World J Surg. Apr 2014; 38(4): 992-6. PMID 24178183
  58. Larsen SG, Wiig JN, Dueland S, et al. Prognostic factors after preoperative irradiation and surgery for locally advanced rectal cancer. Eur J Surg Oncol. Apr 2008; 34(4): 410-7. PMID 17614249
  59. Lim SB, Yu CS, Hong YS, et al. Long-term outcomes in patients with locally advanced rectal cancer treated with preoperative chemoradiation followed by curative surgical resection. J Surg Oncol. Nov 2012; 106(6): 659-66. PMID 22674581
  60. Mannaerts GH, Martijn H, Crommelin MA, et al. Feasibility and first results of multimodality treatment, combining EBRT, extensive surgery, and IOERT in locally advanced primary rectal cancer. Int J Radiat Oncol Biol Phys. May 01 2000; 47(2): 425-33. PMID 10802370
  61. 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. Mar 2008; 393(2): 173-80. PMID 18172677
  62. Mathis KL, Larson DW, Dozois EJ, et al. Outcomes following surgery without radiotherapy for rectal cancer. Br J Surg. Jan 2012; 99(1): 137-43. PMID 22052336
  63. Nakfoor BM, Willett CG, Shellito PC, et al. The impact of 5-fluorouracil and intraoperative electron beam radiation therapy on the outcome of patients with locally advanced primary rectal and rectosigmoid cancer. Ann Surg. Aug 1998; 228(2): 194-200. PMID 9712564
  64. 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. Jan 01 2004; 58(1): 106-12. PMID 14697427
  65. Pacelli F, Di Giorgio A, Papa V, et al. Preoperative radiotherapy combined with intraoperative radiotherapy improve results of total mesorectal excision in patients with T3 rectal cancer. Dis Colon Rectum. Feb 2004; 47(2): 170-9. PMID 15043286
  66. Palmer G, Martling A, Cedermark B, et al. A population-based study on the management and outcome in patients with locally recurrent rectal cancer. Ann Surg Oncol. Feb 2007; 14(2): 447-54. PMID 17139457
  67. Park JH, Yoon SM, Yu CS, et al. Randomized phase 3 trial comparing preoperative and postoperative chemoradiotherapy with capecitabine for locally advanced rectal cancer. Cancer. Aug 15 2011; 117(16): 3703-12. PMID 21328328
  68. Ratto C, Valentini V, Morganti AG, et al. Combined-modality therapy in locally advanced primary rectal cancer. Dis Colon Rectum. Jan 2003; 46(1): 59-67. PMID 12544523
  69. 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. Apr 01 2007; 67(5): 1381-8. PMID 17275208
  70. 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. Sep 2004; 30(7): 750-8. PMID 15296989
  71. Sanfilippo NJ, Crane CH, Skibber J, et al. T4 rectal cancer treated with preoperative chemoradiation to the posterior pelvis followed by multivisceral resection: patterns of failure and limitations of treatment. Int J Radiat Oncol Biol Phys. Sep 01 2001; 51(1): 176-83. PMID 11516868
  72. Sauer R, Becker H, Hohenberger W, et al. Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med. Oct 21 2004; 351(17): 1731-40. PMID 15496622
  73. 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. May 2009; 145(5): 486-94. PMID 19375606
  74. Willett CG, Shellito PC, Tepper JE, et al. Intraoperative electron beam radiation therapy for recurrent locally advanced rectal or rectosigmoid carcinoma. Cancer. Mar 15 1991; 67(6): 1504-8. PMID 2001537
  75. Zhang Q, Tey J, Yang Z, et al. Intraoperative radiotherapy in the combination of adjuvant chemotherapy for the treatment of pT3N0M0 rectal cancer after radical surgery. Am J Clin Oncol. Feb 2014; 37(1): 8-12. PMID 22892433
  76. Zhang Q, Tey J, Yang Z, et al. Adjuvant chemoradiation plus intraoperative radiotherapy versus adjuvant chemoradiation alone in patients with locally advanced rectal cancer. Am J Clin Oncol. Feb 2015; 38(1): 11-6. PMID 25616201
  77. Abuchaibe O, Calvo FA, Azinovic I, et al. Intraoperative radiotherapy in locally advanced recurrent colorectal cancer. Int J Radiat Oncol Biol Phys. Aug 01 1993; 26(5): 859-67. PMID 8344855
  78. Bedrosian I, Giacco G, Pederson L, et al. Outcome after curative resection for locally recurrent rectal cancer. Dis Colon Rectum. Feb 2006; 49(2): 175-82. PMID 16392024
  79. 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. Jul 2008; 15(7): 1937-47. PMID 18389321
  80. Eble MJ, Lehnert T, Treiber M, et al. Moderate dose intraoperative and external beam radiotherapy for locally recurrent rectal carcinoma. Radiother Oncol. Nov 1998; 49(2): 169-74. PMID 10052883
  81. 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. Jan 01 2011; 79(1): 143-50. PMID 20395067
  82. Hansen MH, Balteskard L, Dørum LM, et al. Locally recurrent rectal cancer in Norway. Br J Surg. Oct 2009; 96(10): 1176-82. PMID 19787766
  83. Hashiguchi Y, Sekine T, Sakamoto H, et al. Intraoperative irradiation after surgery for locally recurrent rectal cancer. Dis Colon Rectum. Jul 1999; 42(7): 886-93; discussion 893-5. PMID 10411435
  84. Hashiguchi Y, Sekine T, Kato S, et al. Indicators for surgical resection and intraoperative radiation therapy for pelvic recurrence of colorectal cancer. Dis Colon Rectum. Jan 2003; 46(1): 31-9. PMID 12544519
  85. Kanemitsu Y, Hirai T, Komori K, et al. Prediction of residual disease or distant metastasis after resection of locally recurrent rectal cancer. Dis Colon Rectum. May 2010; 53(5): 779-89. PMID 20389212
  86. Lee JH, Kim DY, Kim SY, et al. Clinical outcomes of chemoradiotherapy for locally recurrent rectal cancer. Radiat Oncol. May 20 2011; 6: 51. PMID 21595980
  87. Lindel K, Willett CG, Shellito PC, et al. Intraoperative radiation therapy for locally advanced recurrent rectal or rectosigmoid cancer. Radiother Oncol. Jan 2001; 58(1): 83-7. PMID 11165686
  88. Mannaerts GH, Rutten HJ, Martijn H, et al. Comparison of intraoperative radiation therapy-containing multimodality treatment with historical treatment modalities for locally recurrent rectal cancer. Dis Colon Rectum. Dec 2001; 44(12): 1749-58. PMID 11742155
  89. Martínez-Monge R, Nag S, Martin EW. Three different intraoperative radiation modalities (electron beam, high-dose-rate brachytherapy, and iodine-125 brachytherapy) in the adjuvant treatment of patients with recurrent colorectal adenocarcinoma. Cancer. Jul 15 1999; 86(2): 236-47. PMID 10421259
  90. Mohiuddin M, Lingareddy V, Rakinic J, et al. Reirradiation for rectal cancer and surgical resection after ultra high doses. Int J Radiat Oncol Biol Phys. Dec 01 1993; 27(5): 1159-63. PMID 8262842
  91. Park JK, Kim YW, Hur H, et al. Prognostic factors affecting oncologic outcomes in patients with locally recurrent rectal cancer: impact of patterns of pelvic recurrence on curative resection. Langenbecks Arch Surg. Jan 2009; 394(1): 71-7. PMID 18663464
  92. Pezner RD, Chu DZ, Ellenhorn JD. Intraoperative radiation therapy for patients with recurrent rectal and sigmoid colon cancer in previously irradiated fields. Radiother Oncol. Jul 2002; 64(1): 47-52. PMID 12208575
  93. Rahbari NN, Ulrich AB, Bruckner T, et al. Surgery for locally recurrent rectal cancer in the era of total mesorectal excision: is there still a chance for cure?. Ann Surg. Mar 2011; 253(3): 522-33. PMID 21209587
  94. Salo JC, Paty PB, Guillem J, et al. Surgical salvage of recurrent rectal carcinoma after curative resection: a 10-year experience. Ann Surg Oncol. Mar 1999; 6(2): 171-7. PMID 10082043
  95. Shoup M, Guillem JG, Alektiar KM, et al. Predictors of survival in recurrent rectal cancer after resection and intraoperative radiotherapy. Dis Colon Rectum. May 2002; 45(5): 585-92. PMID 12004205
  96. Suzuki K, Gunderson LL, Devine RM, et al. Intraoperative irradiation after palliative surgery for locally recurrent rectal cancer. Cancer. Feb 15 1995; 75(4): 939-52. PMID 7531113
  97. 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. Dec 15 1999; 86(12): 2612-24. PMID 10594856
  98. Vermaas M, Nuyttens JJ, Ferenschild FT, et al. Reirradiation, surgery and IORT for recurrent rectal cancer in previously irradiated patients. Radiother Oncol. Jun 2008; 87(3): 357-60. PMID 18353474
  99. Wells BJ, Stotland P, Ko MA, et al. Results of an aggressive approach to resection of locally recurrent rectal cancer. Ann Surg Oncol. Feb 2007; 14(2): 390-5. PMID 17063304
  100. Wiig JN, Poulsen JP, Tveit KM, et al. Intra-operative irradiation (IORT) for primary advanced and recurrent rectal cancer. a need for randomised studies. Eur J Cancer. May 2000; 36(7): 868-74. PMID 10785591
  101. Wiig JN, Larsen SG, Dueland S, et al. Preoperative irradiation and surgery for local recurrence of rectal and rectosigmoid cancer. Prognostic factors with regard to survival and further local recurrence. Colorectal Dis. Jan 2008; 10(1): 48-57. PMID 18028472

Coding Section

Codes Number Description
CPT 77424 Intraoperative radiation treatment delivery, X-ray, single treatment session
  77425 Intraoperative radiation treatment delivery, electrons, single treatment session
  77469 Intraoperative radiation treatment management
  0735T Preparation of tumor cavity, with placement of a radiation therapy applicator for intraoperative radiation therapy (IORT) concurrent with primary craniotomy (List separately in addition to code for primary procedure)
ICD-10-CM C16.0-C16.9 Malignant neoplasm of stomach code range
  C20 Malignant neoplasm of rectum
  C25.0- C25.9 Malignant neoplasm of Pancreas code range
  C51- C58 Malignant neoplasm of female organs code range
  C64.1 -C64.9 Malignant neoplasm of kidney code range
  C7A.026 Malignant carcinoid tumor of the rectum
  C71.0-C71.9 Malignant neoplasm of brain code range
  C74.90, C74.91, C74.92 Malignant neoplasm of unspecified part of adrenal glands
  C78.5 Secondary neoplasm of large intestine and rectum
  C78.80-C78.89 Secondary neoplasm of other and unspecified digestive organs
  D00.2 Carcinoma in-situ of stomach
  D49.2 Neoplasm of unspecified behavior of bone, soft tissue and skin
  D49.8 Neoplasm of unspecified behavior of other sites
ICD-10-PCS   ICD-10-PCS codes are only used for inpatient services.
  DD073Z0 Radiation oncology, gastrointestinal system, beam radiation, rectum, electrons, intraoperative
Type of Service Therapy/Radiation  
Place of Service Inpatient

Procedure and diagnosis codes on Medical Policy documents are included only as a general reference tool for each policy. They may not be all-inclusive.

This medical policy was developed through consideration of peer-reviewed medical literature generally recognized by the relevant medical community, U.S. FDA approval status, nationally accepted standards of medical practice and accepted standards of medical practice in this community, Blue Cross Blue Shield Association technology assessment program (TEC) and other nonaffiliated technology evaluation centers, reference to federal regulations, other plan medical policies and accredited national guidelines.

"Current Procedural Terminology © American Medical Association. All Rights Reserved" 

History From 2013 Forward     

12/14/2023 Annual review, no change to policy intent. Updating coding, rationale and references.
12/01/2022 Annual review, no change to policy intent. Updating background, rationale and references.
09/23/2022 Corrected spelling error in coding section

12/01/2021 

Annual review, no change to policy intent. Updating rationale and references. 

12/01/2020 

Annual review, no change to policy intent. Updating rationale and references. 

12/02/2019 

Annual review, no change to policy intent. Updating rationale and references. 

12/31/2018

Annual review, no change to policy intent. Updating background, description, rationale and references. 

12/13/2017 

Annual review, no change to policy intent. Updating background, description, rationale and references. 

12/01/2016 

Annual review, expanding medical necessity to include: colorectal cancer, pancreatic cancer, pelvic cancers (for example, cervical or uterine), soft tissue sarcomas, updating coding. 

12/2/2015 

Annual review, no change to policy intent. Updating background, description, rationale and references.

12/01/2014 

Annual review. Added coding, regulatory status, policy guidelines, benefit applications. Updated description, background, rationale and references. No change to policy intent.

12/3/2013

Annual review. Added related policies and updated references. 

Complementary Content
${loading}