Skip to main content
Journal of Clinical Oncology logoLink to Journal of Clinical Oncology
. 2014 Mar 17;32(12):1210–1217. doi: 10.1200/JCO.2013.53.6524

Risk Factors for GI Adverse Events in a Phase III Randomized Trial of Bevacizumab in First-Line Therapy of Advanced Ovarian Cancer: A Gynecologic Oncology Group Study

Robert A Burger 1,, Mark F Brady 1, Michael A Bookman 1, Bradley J Monk 1, Joan L Walker 1, Howard D Homesley 1, Jeffrey Fowler 1, Benjamin E Greer 1, Matthew Boente 1, Gini F Fleming 1, Peter C Lim 1, Stephen C Rubin 1, Noriyuki Katsumata 1, Sharon X Liang 1
PMCID: PMC3986384  PMID: 24637999

Abstract

Purpose

To evaluate risk factors for GI adverse events (AEs) within a phase III trial of bevacizumab in first-line ovarian cancer therapy.

Patients and Methods

Women with previously untreated advanced disease after surgery were randomly allocated to six cycles of platinum-taxane chemotherapy plus placebo cycles (C)2 to C22 (R1); chemotherapy plus bevacizumab C2 to C6 plus placebo C7 to C22 (R2); or chemotherapy plus bevacizumab C2 to C22 (R3). Patients were evaluated for history or on-study development of potential risk factors for GI AEs defined as grade ≥ 2 perforation, fistula, necrosis, or hemorrhage.

Results

Of 1,873 patients enrolled, 1,759 (94%) were evaluable, and 2.8% (50 of 1,759) experienced a GI AE: 10 of 587 (1.7%, R1), 20 of 587 (3.4%, R2), and 20 of 585 (3.4%, R3). Univariable analyses indicated that previous treatment of inflammatory bowel disease (IBD; P = .005) and small bowel resection (SBR; P = .032) or large bowel resection (LBR; P = .012) at primary surgery were significantly associated with a GI AE. The multivariable estimated relative odds of a GI AE were 13.4 (95% CI, 3.44 to 52.3; P < .001) for IBD; 2.05 (95% CI, 1.09 to 3.88; P = .026) for LBR; 1.95 (95% CI, 0.894 to 4.25; P = .093) for SBR; and 2.15 for bevacizumab exposure (aggregated 95% CI, 1.05 to 4.40; P = .036).

Conclusion

History of treatment for IBD, and bowel resection at primary surgery, increase the odds of GI AEs in patients receiving first-line platinum-taxane chemotherapy for advanced ovarian cancer. After accounting for these risk factors, concurrent bevacizumab doubles the odds of a GI AE, but is not appreciably increased by continuation beyond chemotherapy.

INTRODUCTION

Angiogenesis is a process integral to disease progression for solid tumors including ovarian cancer and is largely promoted by vascular endothelial growth factor (VEGF).117 Bevacizumab, a VEGF neutralizing monoclonal antibody,18 has demonstrated single agent activity in phase II ovarian cancer trials.19,20 Results of four phase III trials have been reported, all demonstrating significant prolongation of progression-free survival when bevacizumab was combined with and continued beyond standard chemotherapy.2124

The incorporation of bevacizumab into first-line ovarian cancer therapy has been controversial because of lack of an overall survival benefit as yet demonstrated for the entire study populations in the two first-line phase III trials22,23 and to concerns related to additional toxicity.25 GI wall disruption is perhaps the most concerning adverse effect associated with bevacizumab and has been reported in approximately 2.4% in general.25 In the phase III first-line ovarian cancer trials, the aggregate rate was 2.9% for 1,960 women allocated to receive bevacizumab and 1.7% for 1,354 in the control groups. The pathogenesis for this complication in the setting of bevacizumab therapy remains unclear, and specific risk factors have been suggested only through historical studies. Therefore, the Gynecologic Oncology Group (GOG) conducted a preplanned study embedded within its first-line phase III trial.

PATIENTS AND METHODS

As shown in Figure 1, GOG 021822 was a double-blind, placebo controlled first-line phase III clinical trial in which women with advanced cancers were randomly allocated to one of three postoperative regimens: six cycles of intravenous carboplatin-paclitaxel chemotherapy cycles (C) 2 to C22 (R1); chemotherapy plus bevacizumab (15 mg/kg) C2 to C6 (R2); and chemotherapy plus bevacizumab C2 to C22 (R3). Participation required stage III incompletely resectable intra-abdominal disease or stage IV disease and a GOG performance status (PS) of 0 to 2. Because of concerns regarding the risk of GI perforation, patients with evidence of intestinal obstruction requiring parenteral hydration or nutrition were excluded. Safety was monitored through physical and laboratory assessment after each treatment cycle by using National Cancer Institute Common Toxicity Criteria version 3.

Fig 1.

Fig 1.

CONSORT diagram. AUC, area under the curve; FIGO, International Federation of Gynecology and Obstetrics staging system.

The presence or absence of potential baseline risk factors for the development of GI adverse events (AEs) including surgical, vascular, hematologic, and inflammatory conditions was collected on a medical history (MEDH) form (online only). Other putative risk factors derived from the database at the completion of the trial included age at enrollment; GOGPS; the combination of stage and debulking level; time from surgery to C1 treatment; time from surgery to C2 treatment; and on-study development of intestinal obstruction, complicated (febrile or grade 4) neutropenia, or thromboembolic events.

GI AEs were defined as grade ≥ 2 perforation, fistula, necrosis, or hemorrhage occurring as of C2 (when bevacizumab or placebo was initiated) and ≤ 30 days of last protocol treatment.

The primary analysis of the clinical trial was based on progression-free survival and was conducted after 423 disease progression events or deaths had been reported. The current study is based on data collected up to the time of the primary data freeze, including only those with collected MEDH forms. Univariable analyses to test the association of GI AEs with potential risk factors were performed by using Fisher's exact test for discrete variables and a logistic model for continuous variables.26 A logistic model was used to estimate the relative odds of a GI AE event because of bevacizumab, adjusted for other risk factors.27 A time-dependent proportional hazards model was used to evaluate the null hypothesis that the onset of febrile neutropenia, intestinal obstruction, or thromboembolic events was not associated with subsequent onset of a GI AE.28 All P values are two-sided.

RESULTS

Patient Characteristics

Baseline characteristics of the 1,873 patients enrolled onto GOG 0218 were similarly distributed across treatment groups (Table 1). The median age was 60 years, over 80% had serous adenocarcinomas, and the majority of cancers were grade 3. Forty percent had stage III disease with residual intra-abdominal tumor implants > 1 cm in diameter after primary cytoreductive surgery, and 26% had stage IV disease.

Table 1.

Baseline Characteristics of the Source Population

Characteristic R1 (n = 625)
R2 (n = 625)
R3 (n = 623)
No. % No. % No. %
Age
    Median 60 60 60
    Range 25-86 24-88 22-89
Race*
    Non-Hispanic white 526 84 519 83 521 84
    Asian 41 7 37 6 39 6
    Non-Hispanic black 25 4 28 5 27 4
    Hispanic 21 3 28 5 25 4
    Other, specified 8 1 5 < 1 4 < 1
GOG PS
    0 311 50 315 50 305 49
    1 272 44 270 43 267 43
    2 42 7 40 6 51 8
Stage surgical tumor debulking level
    III (macro) ≤ 1 cm 218 35 205 33 216 35
    III > 1 cm§ 254 41 256 41 242 39
    IV 153 25 164 26 165 27
Histology
    Serous 541 87 519 83 524 84
    Endometrioid 21 3 14 2 24 4
    Clear cell 12 2 23 4 20 3
    Mucinous 6 1 5 < 1 8 1
    Other/not specified 45 7 64 10 47 7
Tumor grade
    3 445 71 465 74 460 74
    2 102 16 86 14 97 16
    1 36 6 28 4 18 3
    Not specified 42 7 46 7 48 8

NOTE. All clear cell tumors classified grade 3.

Abbreviations: GOG, Gynecologic Oncology Group; PS, performance status; R1, chemotherapy plus placebo followed by placebo; R2, chemotherapy + bevacizumab followed by placebo; R3, chemotherapy + bevacizumab followed by bevacizumab.

*

Percentages may not total 100% because of rounding or categorization.

International Federation of Gynecology and Obstetrics.

Stage III with ≤ 1 cm maximal diameter for any residual abdominal tumor.

§

Stage III with > 1 cm maximal diameter disease in any residual abdominal tumor.

Results from central GOG Pathology Committee review updated September 2010.

MEDH forms were collected on 1,759 (94%) of the 1,873 patients enrolled. As shown in Table 2, the frequency of putative medical risk factors for GI AEs was similarly distributed across the three treatment groups. Of note, at least 30% of patients had a vascular risk factor, with 582 (33%) under current medical management for hypertension and 226 (12.8%) having smoked within the previous year. With respect to GI conditions, 147 patients (8.3%) underwent small bowel resection (SBR), and 306 (17.4%) underwent large bowel resection (LBR) at primary surgery for ovarian cancer. Relatively few had pre-existing Crohn disease or ulcerative colitis.

Table 2.

Distribution of Medical History Risk Factors by Randomized Treatment Group

R1 (n = 587)
R2 (n = 587)
R3 (n = 585)
Total (N = 1,759)
No. % No. % No. % No. %
Currently takes medication for hypertension 191 32.5 201 34.2 190 32.5 582 33.0
Has in the past taken medication for hypertension 30 5.1 34 5.8 25 4.3 89 5.1
Ever had peripheral vascular disease with claudication 5 0.8 2 0.3 3 0.5 10 0.5
Ever had myocardial infarct or cerebrovascular accident 11 1.9 16 2.7 15 2.6 42 2.3
Smoked within past year 81 13.8 77 13.1 68 11.6 226 12.8
Currently takes insulin for diabetes 9 1.5 11 1.9 12 2.0 32 1.8
Currently takes oral medication for diabetes 31 5.3 42 7.2 42 7.2 115 6.5
Ever diagnosed with autoimmune disease 16 2.7 15 2.6 15 2.6 46 2.6
Ever had a peptic ulcer 11 1.9 15 2.6 11 1.9 37 2.1
Ever had an intestinal obstruction radiographically 16 2.7 18 3.1 18 3.1 52 3.0
Ever diagnosed with Crohn disease 2 0.3 1 0.2 1 0.2 4 0.2
Ever diagnosed with ulcerative colitis 6 1.0 7 1.2 3 0.5 16 0.9
Ever had medication or surgery for inflammatory bowel disease 7 1.2 3 0.5 3 0.5 13 0.7
Ever had medication, hospitalization, antibiotics, or dietary modification for diverticulitis 8 1.4 16 2.7 12 2.0 36 2.0
Ever had Clostridium difficile colitis 11 1.9 8 1.3 11 1.9 30 1.7
Ever had small bowel resection with anastomosis 53 9.3 51 8.7 54 9.2 158 9.0
Had small bowel resection at primary surgery 48 8.2 48 8.2 51 8.7 147 8.3
Ever had a large bowel resection with anastomosis 95 16.2 113 19.2 114 19.5 322 18.3
Had large bowel resection at primary surgery 90 15.3 108 18.4 108 18.5 306 17.4
Uses corticosteroids for a chronic condition 11 1.9 8 1.4 11 1.9 30 1.7
Uses NSAID chronically 38 6.5 34 5.8 38 6.5 110 6.3

Abbreviations: NSAID, nonsteroidal anti-inflammatory drug; R1, chemotherapy plus placebo followed by placebo; R2, chemotherapy + bevacizumab followed by placebo; R3, chemotherapy + bevacizumab followed by bevacizumab.

The rates of potential risk factors developing after enrollment but during treatment were low, including 39 patients (2.2%) with GI obstruction, 86 (4.9%) with complicated neutropenia (either febrile or grade 4), and 100 (5.7%) with either arterial or venous thromboembolic events.

Incidence and Spectrum of GI AEs

Fifty-seven patients (3.2%) experienced a GI AE overall, and in 50 patients (2.8%), the event occurred during or beyond treatment C2 (start of bevacizumab or placebo). Three patients (one patient with grade 3 hemorrhage, one with grade 3 fistula, and one with grade 5 bowel necrosis) are excluded from this report because for these cases a MEDH case report form was not submitted. As shown in Table 3, the most common type of GI AE was perforation in 20 patients (40%), followed by hemorrhage in 14 (28%) and fistula in 12 (24%). GI AEs were reported in 10 of 587 patients (1.7%), 20 of 587 (3.4%), and 20 of 585 (3.4%) for patients assigned to R1, R2, and R3, respectively. Although there were no grade 4 or 5 events in the R1 treatment group, events classified as life-threatening or fatal occurred in six (1%) in the R2 group and four (0.7%) in the R3 group. Figure 2 illustrates the timing of events as a function of treatment group and cycle. Forty-six (92%) occurred during C2 through C6 (chemotherapy phase), with 38 (75%) reported by C4.

Table 3.

Number of Grade 2 or Worse GI Adverse Event at or Beyond Cycle 2 by Type, Grade, and Treatment Group

GI Adverse Event Type R1
R2
R3
Total
2 3 4 5 2 3 4 5 2 3 4 5
Fistula 1 3 0 0 1 2 1 0 1 3 0 0 12
GI leak 0 0 0 0 0 0 0 0 0 1 2 0 3
Necrosis 0 1 0 0 0 0 0 0 0 0 0 0 1
Perforation 0 2 0 0 1 5 2 3 0 5 0 2 20
Bleeding 2 1 0 0 3 2 0 0 3 3 0 0 14
Total 3 7 0 0 5 9 3 3 4 12 2 2 50*

Abbreviations: R1, chemotherapy plus placebo followed by placebo; R2, chemotherapy + bevacizumab followed by placebo; R3, chemotherapy + bevacizumab followed by bevacizumab.

*

Three patients who experienced a grade 2 or worse GI adverse event after cycle 2 had no medical history form submitted and are therefore not included in this table or the rest of this report.

Fig 2.

Fig 2.

Number of treatment cycles before grade 2 or worse GI adverse event (AE). There were seven GI AEs before treatment cycle 2 that are not included in this figure. R1, chemotherapy plus placebo followed by placebo; R2, chemotherapy + bevacizumab followed by placebo; R3, chemotherapy + bevacizumab followed by bevacizumab.

Association Between Risk Factors and GI AEs

Univariable analysis indicated no association of GI AEs with age at enrollment (P = .593), baseline PS (P = .297), the combination of stage and residual disease after debulking surgery (P = .378), time from surgery to C1 treatment (P = .625), or time from surgery to C2 treatment (P = .997; data not shown). There was also no significant association between a GI AE and on-study development of GI obstruction (P = .625), thromboembolic events (P = .202), or complicated neutropenia (P = .094, online only). In the time-dependent proportional hazards model, the odds of a GI AE was greater among the 74 patients in the study population who experienced febrile neutropenia before the onset of the GI AE, with a hazard ratio of 2.55 (95% confidence limits, 0.607 to 10.74), but this was not statistically significant (P = .201).

As shown in Table 4, a significant association with a GI AE was observed for history of inflammatory bowel disease (IBD; P = .020) or treatment of IBD (P = .005); SBR with anastomosis (P = .039) or SBR at primary surgery (P = .032); and LBR with anastomosis (P = .016) or LBR at primary surgery (P = .012). A multivariable logistic model was used to estimate the odds of a GI AE associated with bevacizumab, adjusted for significantly associated risk factors: IBD treatment, SBR at primary surgery, and LBR at primary surgery. Relative to R1, the odds of a GI AE were 2.15 (95% CI, 0.981 to 4.71) for R2 and 2.15 (95% CI, 0.981 to 4.70) for R3. Because the odds of a GI AE in each of the bevacizumab treated groups relative to placebo were similar, these two groups were combined into a single group to provide a more precise estimate of the effect of bevacizumab relative to placebo. As shown in Table 5, multivariable analysis estimated that bevacizumab independently increases the incidence of a GI AE by 2.15 (95% CI, 1.049 to 4.40; exact P = .032), which is similar to the univariable estimate, 2.19. Previous treatment of IBD and LBR at primary surgery were also independently associated with increased odds, whereas the impact on increased odds for SBR was similar in magnitude to LBR but was not statistically significant (P = .093).

Table 4.

Univariable Analyses of Medical History Risk Factors and Development of a GI Adverse Event

− GI AE (n = 1,709)
+ GI AE (n = 50)
P
No. % No. %
Medication for hypertension .170
    Never 1,063 98 25 2.3
    Past 86 97 3 3.4
    Current 560 96 22 3.8
Peripheral vascular disease with claudication 1.00
    Never 1,699 97 50 2.9
    Ever 10 100 0.0 0.0
Myocardial infarct or cerebrovascular accident .114
    Never 1,670 97 47 2.7
    Ever 39 93 3 7.1
Smoked within past year .829
    No 1,490 97 43 2.8
    Yes 219 97 7 3.1
Current medication for diabetes .909
    No 1,566 97 46 2.9
    Oral 112 97 3 2.6
    Insulin 31 97 1 3.1
Diagnosis of autoimmune disease .379
    Never 1,665 97 48 2.8
    Ever 44 96 2 4.4
Peptic ulcer .624
    Never 1,672 97 50 2.9
    Ever 37 100 0 0.0
Intestinal obstruction radiographically .658
    Never 1,659 97 48 2.8
    Ever 50 96 2 3.9
Diagnosis of IBD .020
    Never 1,692 97 47 2.7
    Crohn disease 4 100 0 0.0
    UC 13 81 3 18.75
Medication or surgery for IBD .005
    Never 1,699 97 47 2.7
    Ever 10 77 3 23
Medication, hospitalization, antibiotics, or dietary modification for diverticulitis 1.000
    Never 1,674 97 49 2.8
    Ever 35 97 1 2.8
Diagnosis of Clostridium difficile colitis .209
    Never 1,681 97 48 2.8
    Ever 28 93 2 6.7
SBR with anastomosis .039
    Never 1,560 97 41 2.6
    Ever 149 94 9 5.7
SBR at primary surgery .032
    No 1,571 97 41 2.5
    Yes 138 94 9 6.1
LBR with anastomosis .016
    Never 1,403 97 34 2.4
    Ever 306 95 16 5.0
LBR at primary surgery .012
    No 1,419 98 34 2.3
    Yes 290 95 16 5.2
Current corticosteroids for a chronic condition .582
    Yes 1,680 97 49 2.8
    No 29 97 1 3.3
Current chronic NSAID use .367
    Yes 1,600 97 49 3.0
    No 109 99 1 0.9

Abbreviations: AE, adverse event; IBD, inflammatory bowel disease; LBR, large bowel resection; NSAID, nonsteroidal anti-inflammatory drug; SBR, small bowel resection; UC, ulcerative colitis.

Table 5.

Results of a Logistic Model Assessing the Association Between GI Adverse Events and Risk Factors Significantly Associated With GI Adverse Events in Univariable Analyses

Model Covariate Relative Odds of Grade ≥ 2 GI AE
P
Estimated Odds 95% CI
IBD treatment 13.40 3.44 to 52.30 < .001
LBR at primary surgery 2.05 1.09 to 3.88 .026
SBR at primary surgery 1.95 0.894 to 4.25 .093
BEV treatment 2.15 1.05 to 4.40 .032

Abbreviations: AE, adverse event; BEV, bevacizumab; IBD, inflammatory bowel disease; LBR, large bowel resection; SBR, small bowel resection.

Finally, a logistic model with a treatment-factor interaction term demonstrated no clear evidence that bevacizumab increased the odds (P > .10) of a GI AE associated with risk factors found to be significant in univariable analyses (data not shown).

DISCUSSION

To our knowledge, this is the first prospective study examining risk factors for serious GI AEs in the context of treatment of solid tumors with anti-VEGF agents.

In the initial report for GOG 0218, GI events were classified as grade ≥ 2 perforation, fistula, necrosis, or leak, and the rates of these events during or after treatment cycle 2 were 1.2%, 2.8%, and 2.6% for those enrolled to R1, R2, and R3, respectively.22 Although one may consider these rates to be relatively low, they should be considered clinically meaningful, as events are often life threatening,29,30 frequently require major surgical intervention, and may have long-term adverse consequences. Thus, it is important that independent and interactive risk factors for these complications are identified, allowing for refined risk assessment, creation of guidelines for patient counseling, development of preventive strategies, and generation of hypotheses related to underlying pathogenic mechanisms.

In the current study, GI AEs also included ≥ grade 2 GI hemorrhage, because mucosal ulceration manifested as bleeding was hypothesized to be mechanistically related to perforation on the basis of pathology studies.31 Using our modified definition for the current study, the rate of GI AEs was 3.4% in each bevacizumab treatment group, approximately double that seen in the group assigned to chemotherapy alone and, not unexpectedly, remained an independent risk factor in multivariable analysis. We examined only events after initiation of bevacizumab or placebo to accurately factor in relative odds attributable to bevacizumab.

The finding that bevacizumab exposure was associated with increased odds of a GI AE with an odds ratio of 2.15 in the current study is supported by a meta-analysis by Hapani et al29 examining 12,294 patients with a variety of nongynecologic solid tumors from 17 randomized controlled trials. This demonstrated the incidence of GI perforation to be 0.9% among patients receiving bevacizumab and the relative odds of GI perforation compared with control patients to be 2.14 (95% CI, 1.19 to 3.85; P = 0.011). Interestingly, in the current study, the odds ratio for the association of bevacizumab exposure with a GI AE was identical (2.15) for both those receiving bevacizumab only during the chemotherapy phase and those receiving bevacizumab during and continued beyond the chemotherapy phase, suggesting that there is no significant additional risk of bevacizumab continued beyond primary platinum-taxane chemotherapy.

The etiology of GI AEs as defined above has been found to be multifactorial and include traumatic, vascular, infectious/inflammatory, or toxic insults to the GI tract with impairment in healing as the common denominator.29 This explains our selection of hypothetical risk factors for investigation. Except for bevacizumab exposure, all risk factors examined were similarly distributed across treatment groups in the clinical trial. We found that the odds of a GI AE was significantly associated with bevacizumab exposure; history of IBD or treatment of IBD; and previous SBR/LBR with anastomosis or SBR/LBR at time of primary surgery before enrolling onto GOG 0218.

Although the mechanisms by which bevacizumab may contribute to development of bowel perforation remain elusive, the preponderance of data suggests impairment of healing at sites of GI injury to be the common denominator. Early preclinical studies before the development of an anti-VEGF monoclonal antibody demonstrated several naturally occurring and synthetic antiangiogenic agents to impair healing after cutaneous surgical trauma32,33 and colonic anastomoses,34,35 whereas others demonstrated no clear impact on healing of cutaneous wounds despite reduction in micro-vascular maturation and granulation tissue.3639 Data pooled from two positive phase III randomized trials of bevacizumab in first-line therapy of metastatic colorectal cancer involving 1,132 patients demonstrated surgical wound healing complications in 1.3% of those receiving bevacizumab versus 0.5% in control patients. Though inconclusive, bevacizumab induced necrosis of tumor invading the bowel wall as an alternative process is suggested by retrospective analyses of clinical,40,41 pathologic,30,42 and radiologic20 data. Indeed, the odds for GI perforation appear to be greater for patients with tumors involving the abdominal cavity, such as colorectal,29 renal cell,29 and ovarian,43 which suggests that this is a possible mechanism. However, one cannot exclude the possibility that such tumors are more likely to be associated with confounding factors, such as tumor infiltration of the mesentery with compromise blood supply, intestinal obstruction, and previous intestinal surgery. In addition, this would not explain the association of such events with bevacizumab in patients with cancer who have no evidence of intra-abdominal tumor and no other obvious risks.29,44 A limitation of the current study is our inability to examine GI wall involvement by tumor prospectively because no reliable methods of diagnosis or documentation have been established. Another possibility is that bevacizumab could limit the blood flow to the splanchnic microvasculature via thrombosis or vasoconstriction,45,46 leading to GI ischemia, but evidence for this hypothesis is limited.

We also found that history of bowel resection was associated with a two- to threefold increase in the odds of a GI AE. The vast majority of GI AEs in this subset of patients is presumed to be due to anastomotic dehiscence, a well-known complication of intestinal surgery in general and most commonly associated with colorectal anastomoses.4749 The risk of anastomotic leak in patients with advanced ovarian carcinoma and related gynecologic malignancies undergoing colorectal surgery may be a particular problem because of protein malnutrition that is common when these surgeries are performed before induction chemotherapy.50 Results of a phase III trial reported by Vergote et al,51 in which women with advanced ovarian cancer were randomly assigned to undergo primary debulking surgery before six cycles of postoperative chemotherapy or to undergo neoadjuvant chemotherapy (NACT) with interval debulking surgery after the third cycle of NACT, demonstrated that the rate of bowel resection at interval debulking surgery after NACT was approximately 50% lower (8.7% of 322 patients) than the rate at primary debulking surgery (15.5% of 310 patients).51 The direct impact on this difference on the development of a GI AE as herein defined is unknown.

An important observation made in the current study is that not only were bevacizumab exposure and previous treatment of IBD and LBR at primary surgery independently associated with an increase in the odds of a GI AE, but also that in a logistic model there was no evidence that bevacizumab exacerbated the risk of a GI AE when these risk factors were present.

The interpretation of our data has additional limitations. First, several variables hypothetically associated with GI AEs were not specifically analyzed, including GI obstruction before treatment (an exclusion criterion for GOG 0218), protein malnutrition, specific chemotherapeutic regimen (identical across treatment groups), or bevacizumab dose density. Unfortunately we did not specifically examine laboratory indices of nutrition, such as pretreatment serum albumin. However, we found no association with PS, which may reflect nutritional state. Cytotoxic regimens including taxanes have been implicated, with mechanisms including necrosis of invasive tumor, bowel wall cytotoxicity, or transmigration of bacteria secondary to neutropenia.5255 The association of bevacizumab dose density with risk of GI AEs has been suggested in the meta-analysis by Hapani et al29; however, these were univariable analyses unadjusted for other factors such as type of combined chemotherapeutic agents. GOG 0218 utilized only one dose density for bevacizumab −5 mg/kg per week. The results of an open label phase III trial of bevacizumab in first-line therapy consisting of the same chemotherapy regimen utilized in GOG 0218 but with a bevacizumab dose density of 2.5 mg/kg per week demonstrated a similar frequency of GI AEs.23 Thus far there is no evidence of a relationship to bevacizumab dose density in the management of patients with ovarian cancer.

Second, type I and type II statistical error could have influenced results and conclusions. Some positive findings could possibly be due to unadjusted multiple hypothesis testing. IBDs are rare conditions associated with development of GI perforation and fistula (particularly for Crohn disease).5658 In the current study, though a history of IBD or treatment thereof was reported in < 1% of the study population, those with a history of treatment for IBD had a 13-fold higher odds of a GI AE compared with those without such history.

The more concerning issue relates to negative findings where no statistically significant associations were found between biologically plausible risk factors and GI AEs. There are several possible explanations, including the relatively low number of events and the low frequency of some risk factors, creating imprecision in risk estimates. Arguably, examples of this phenomenon may be on-study development of GI obstruction or complicated neutropenia, occurring in 39 (2.2%) and 86 (4.9%), respectively. Intestinal obstruction is a well-established risk factor for GI perforation59 and has retrospectively been found to be a risk factor in a registry of approximately 2,000 patients with metastatic colorectal cancer treated with bevacizumab.41 Severe neutropenia is thought to permit trans-mural invasion of pathogenic bacteria and has been implicated as a risk factor for enterocolonic perforation.60

Third, our results may not be completely applicable to other primary treatment scenarios, such as for patients with no macroscopic residual disease after initial surgery, receiving alternative regimens such as dose-dense paclitaxel with carboplatin,61 treated with combined intraperitoneal-intravenous chemotherapy, or receiving induction NACT with interval surgical cytoreduction. Furthermore, the results may be even less applicable to those receiving bevacizumab with or without chemotherapy in the recurrent disease setting.

On the basis of our findings, we conclude that history of treatment for IBD and colon resection during initial surgery for advanced ovarian cancer appears to increase the risk of bowel perforation, fistula, necrosis, or bleeding during first-line platinum-taxane based treatment. After accounting for these risk factors, first-line treatment with bevacizumab also increases the odds of such a GI event, but bevacizumab does not appear to synergize with these other risk factors or increase the odds of GI AEs when it is continued after completion of platinum-taxane primary chemotherapy. Patients preparing for first-line therapy after surgery should be carefully counseled regarding the potential impact of these interventions and conditions on their individual risks.

Supplementary Material

Protocol

Acknowledgment

We thank Suzanne Baskerville and Kristin Engel, GOG Statistical and Data Center (SDC), Buffalo, NY, for their support in data coordination; Anne Reardon, GOG SDC, and the GOG Publications Subcommittee for their assistance in manuscript preparation and review, respectively.

Glossary Terms

angiogenesis:

the process involved in the generation of new blood vessels. Although this is a normal process that naturally occurs and is controlled by so-called on and off switches, blocking tumor angiogenesis (antiangiogenesis) disrupts the blood supply to tumors, thereby preventing tumor growth.

bevacizumab:

also called Avastin (Genentech, South San Francisco, CA). Bevacizumab is a recombinant, humanized, monoclonal antibody that binds and neutralizes the vascular endothelial growth factor, thus acting as an antiangiogenic agent.

proportional hazards:

semiparametric approach to survival analysis developed by Cox in 1972. Unlike product-limit (Kaplan-Meier) survival analyses, which are restricted to categorical predictor variables and do not produce a risk estimate, proportional hazards models can accommodate continuous and ordinal variables as well as allow for the inclusion of multiple predictor variables to compute adjusted risk estimates. Proportional hazards models are based on the fundamental premise that all individuals have the same baseline hazard that varies as a function of time [(t)], but that exposure to the independent variable changes the hazard by a fixed value [h(x)]. What is parameterized in the model is the value of this fixed effect per unit increase of the predictor variable whereas the value of (t) remains uncharacterized.

Appendix

The following institutions participated in the study: Abington Memorial Hospital, Abramson Cancer Center at the University of Pennsylvania, Aurora Women's Pavilion of West Allis Memorial Hospital, Cleveland Clinic Foundation, Community Clinical Oncology Program, Cooper Hospital University Medical Center, CTSU, Duke University Medical Center, Fox Chase Cancer Center, Fred Hutchinson Cancer Research Center, Georgia Core, Gynecologic Oncology Network, Gynecologic Oncology of West Michigan, PLLC, Indiana University Medical Center, MD Anderson Cancer Center, Magee Women's Hospital–University of Pittsburgh Medical Center, Mayo Clinic Rochester, Memorial Sloan-Kettering Cancer Center, Moffitt Cancer Center and Research Institute, Mount Sinai Medical Center, New York University Medical Center, Northwestern University, The Ohio State University Medical Center, Penn State Milton S. Hershey Medical Center, Roswell Park Cancer Institute, Rush University Medical Center, Saitama Medical University International/GOG Japan, Seoul National University Hospital/KGOG, State University of New York Downstate Medical Center, Stony Brook University Medical Center, The Hospital of Central Connecticut, University Hospitals–Ireland Cancer Center, University of Alabama at Birmingham, University of California at Los Angeles, University of California Medical Center at Irvine–Orange Campus, University of Chicago, University of Cincinnati, University of Colorado Cancer Center–Anschutz Cancer Pavilion, University of Iowa Hospitals and Clinics, University of Kentucky, University of Massachusetts Medical School, University of Minnesota Medical Center–Fairview, University of Mississippi Medical Center, University of New Mexico, University of North Carolina, University of Oklahoma Health Sciences Center, University of Texas Medical Branch University of Texas Southwestern Medical Center, University of Virginia, University of Wisconsin Hospitals and Clinics, Wake Forest University Health Sciences, Walter Reed Army Medical Center, Washington University School of Medicine, Wayne State University, Women and Infants' Hospital, Women's Cancer Center of Nevada and Yale University.

Footnotes

Listen to the podcast by Dr Markman at www.jco.org/podcasts

Supported by National Cancer Institute grants to the Gynecologic Oncology Group Administrative Office (CA 27469) and the Gynecologic Oncology Group Statistical and Data Center (CA 37517).

Terms in blue are defined in the glossary, found at the end of this article and online at www.jco.org.

Presented in part at the 2011 Meeting of the Society of Gynecologic Oncologists, March 2011, Orlando, FL.

Authors' disclosures of potential conflicts of interest and author contributions are found at the end of this article.

Clinical trial information: NCT00262847.

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

The author(s) indicated no potential conflicts of interest.

AUTHOR CONTRIBUTIONS

Final approval of manuscript: All authors

REFERENCES

  • 1.Byrne AT, Ross L, Holash J, et al. Vascular endothelial growth factor-trap decreases tumor burden, inhibits ascites, and causes dramatic vascular remodeling in an ovarian cancer model. Clin Cancer Res. 2003;9:5721–5728. [PubMed] [Google Scholar]
  • 2.Chen H, Ye D, Xie X, et al. VEGF, VEGFRs expressions and activated STATs in ovarian epithelial carcinoma. Gynecol Oncol. 2004;94:630–635. doi: 10.1016/j.ygyno.2004.05.056. [DOI] [PubMed] [Google Scholar]
  • 3.Manenti L, Riccardi E, Marchini S, et al. Circulating plasma vascular endothelial growth factor in mice bearing human ovarian carcinoma xenograft correlates with tumor progression and response to therapy. Mol Cancer Ther. 2005;4:715–725. doi: 10.1158/1535-7163.MCT-04-0305. [DOI] [PubMed] [Google Scholar]
  • 4.Belotti D, Calcagno C, Garofalo A, et al. Vascular endothelial growth factor stimulates organ-specific host matrix metalloproteinase-9 expression and ovarian cancer invasion. Mol Cancer Res. 2008;6:525–534. doi: 10.1158/1541-7786.MCR-07-0366. [DOI] [PubMed] [Google Scholar]
  • 5.Trinh XB, Tjalma WA, Vermeulen PB, et al. The VEGF pathway and the AKT/mTOR/p70S6K1 signalling pathway in human epithelial ovarian cancer. Br J Cancer. 2009;100:971–978. doi: 10.1038/sj.bjc.6604921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Goodheart MJ, Ritchie JM, Rose SL, et al. The relationship of molecular markers of p53 function and angiogenesis to prognosis of stage I epithelial ovarian cancer. Clin Cancer Res. 2005;11:3733–3742. doi: 10.1158/1078-0432.CCR-04-0056. [DOI] [PubMed] [Google Scholar]
  • 7.Hollingsworth HC, Kohn EC, Steinberg SM, et al. Tumor angiogenesis in advanced stage ovarian carcinoma. Am J Pathol. 1995;147:33–41. [PMC free article] [PubMed] [Google Scholar]
  • 8.Raspollini MR, Amunni G, Villanucci A, et al. Prognostic significance of microvessel density and vascular endothelial growth factor expression in advanced ovarian serous carcinoma. Int J Gynecol Cancer. 2004;14:815–823. doi: 10.1111/j.1048-891X.2004.014514.x. [DOI] [PubMed] [Google Scholar]
  • 9.Paley PJ, Staskus KA, Gebhard K, et al. Vascular endothelial growth factor expression in early stage ovarian carcinoma. Cancer. 1997;80:98–106. doi: 10.1002/(sici)1097-0142(19970701)80:1<98::aid-cncr13>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
  • 10.Gasparini G, Bonoldi E, Viale G, et al. Prognostic and predictive value of tumour angiogenesis in ovarian carcinomas. Int J Cancer. 1996;69:205–211. doi: 10.1002/(SICI)1097-0215(19960621)69:3<205::AID-IJC10>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
  • 11.Siddiqui GK, Elmasry K, Wong Te Fong AC, et al. Prognostic significance of intratumoral vascular endothelial growth factor as a marker of tumour angiogenesis in epithelial ovarian cancer. Eur J Gynaecol Oncol. 2010;31:156–159. [PubMed] [Google Scholar]
  • 12.Smerdel MP, Waldstrom M, Brandslund I, et al. Prognostic importance of vascular endothelial growth factor-A expression and vascular endothelial growth factor polymorphisms in epithelial ovarian cancer. Int J Gynecol Cancer. 2009;19:578–584. doi: 10.1111/IGC.0b013e3181a13168. [DOI] [PubMed] [Google Scholar]
  • 13.Alvarez AA, Krigman HR, Whitaker RS, et al. The prognostic significance of angiogenesis in epithelial ovarian carcinoma. Clin Cancer Res. 1999;5:587–591. [PubMed] [Google Scholar]
  • 14.Shen GH, Ghazizadeh M, Kawanami O, et al. Prognostic significance of vascular endothelial growth factor expression in human ovarian carcinoma. Br J Cancer. 2000;83:196–203. doi: 10.1054/bjoc.2000.1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Duncan TJ, Al-Attar A, Rolland P, et al. Vascular endothelial growth factor expression in ovarian cancer: A model for targeted use of novel therapies? Clin Cancer Res. 2008;14:3030–3035. doi: 10.1158/1078-0432.CCR-07-1888. [DOI] [PubMed] [Google Scholar]
  • 16.Mesiano S, Ferrara N, Jaffe RB. Role of vascular endothelial growth factor in ovarian cancer: Inhibition of ascites formation by immunoneutralization. Am J Pathol. 1998;153:1249–1256. doi: 10.1016/S0002-9440(10)65669-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Xu L, Yoneda J, Herrera C, et al. Inhibition of malignant ascites and growth of human ovarian carcinoma by oral administration of a potent inhibitor of the vascular endothelial growth factor receptor tyrosine kinases. Int J Oncol. 2000;16:445–454. doi: 10.3892/ijo.16.3.445. [DOI] [PubMed] [Google Scholar]
  • 18.Presta LG, Chen H, O'Connor SJ, et al. Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Res. 1997;57:4593–4599. [PubMed] [Google Scholar]
  • 19.Burger RA, Sill MW, Monk BJ, et al. Phase II trial of bevacizumab in persistent or recurrent epithelial ovarian cancer or primary peritoneal cancer: A Gynecologic Oncology Group Study. J Clin Oncol. 2007;25:5165–5171. doi: 10.1200/JCO.2007.11.5345. [DOI] [PubMed] [Google Scholar]
  • 20.Cannistra SA, Matulonis UA, Penson RT, et al. Phase II study of bevacizumab in patients with platinum-resistant ovarian cancer or peritoneal serous cancer. J Clin Oncol. 2007;25:5180–5186. doi: 10.1200/JCO.2007.12.0782. [DOI] [PubMed] [Google Scholar]
  • 21.Aghajanian C, Blank SV, Goff BA, et al. OCEANS: A randomized, double-blind, placebo-controlled phase III trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent epithelial ovarian, primary peritoneal, or fallopian tube cancer. J Clin Oncol. 2012;30:2039–2045. doi: 10.1200/JCO.2012.42.0505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Burger RA, Brady MF, Bookman MA, et al. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med. 2011;365:2473–2483. doi: 10.1056/NEJMoa1104390. [DOI] [PubMed] [Google Scholar]
  • 23.Perren TJ, Swart AM, Pfisterer J, et al. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med. 2011;365:2484–2496. doi: 10.1056/NEJMoa1103799. [DOI] [PubMed] [Google Scholar]
  • 24.Pujade-Lauraine EH F, Weber B, Reuss A, et al. AURELIA: A randomized phase III trial evaluating bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer. J Clin Oncol. 2012;30:327s. doi: 10.1200/JCO.2013.51.4489. (abstr LBA5002) [DOI] [PubMed] [Google Scholar]
  • 25.Avastin. Full Prescribing Information 2013. http://www.gene.com/download/pdf/avastin_prescribing.pdf.
  • 26.Cox DR. The Analysis of Binary Data. London, UK: Chapman and Hall; 1970. [Google Scholar]
  • 27.Hosmer DWL. Applied Logistic Regression. 2nd ed. New York, NY: John Wiley and Sons; 2000. [Google Scholar]
  • 28.Cox DR. Analysis of Survival Data. London, UK: Chapman and Hall; 1984. [Google Scholar]
  • 29.Hapani S, Chu D, Wu S. Risk of gastrointestinal perforation in patients with cancer treated with bevacizumab: A meta-analysis. Lancet Oncol. 2009;10:559–568. doi: 10.1016/S1470-2045(09)70112-3. [DOI] [PubMed] [Google Scholar]
  • 30.Diaz JP, Tew WP, Zivanovic O, et al. Incidence and management of bevacizumab-associated gastrointestinal perforations in patients with recurrent ovarian carcinoma. Gynecol Oncol. 2010;116:335–339. doi: 10.1016/j.ygyno.2009.11.017. [DOI] [PubMed] [Google Scholar]
  • 31.Tol J, Cats A, Mol L, et al. Gastrointestinal ulceration as a possible side effect of bevacizumab which may herald perforation. Invest New Drugs. 2008;26:393–397. doi: 10.1007/s10637-008-9125-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Klein SA, Bond SJ, Gupta SC, et al. Angiogenesis inhibitor TNP-470 inhibits murine cutaneous wound healing. J Surg Res. 1999;82:268–274. doi: 10.1006/jsre.1998.5551. [DOI] [PubMed] [Google Scholar]
  • 33.Streit M, Velasco P, Riccardi L, et al. Thrombospondin-1 suppresses wound healing and granulation tissue formation in the skin of transgenic mice. EMBO J. 2000;19:3272–3282. doi: 10.1093/emboj/19.13.3272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Garcia-Olmo DC, Paya J, Garcia-Olmo D. Effects of perioperative treatment with TNP-470 on the resistance of colonic anastomoses in rats. Dig Surg. 2000;17:154–159. doi: 10.1159/000018820. [DOI] [PubMed] [Google Scholar]
  • 35.te Velde EA, Voest EE, van Gorp JM, et al. Adverse effects of the antiangiogenic agent angiostatin on the healing of experimental colonic anastomoses. Ann Surg Oncol. 2002;9:303–309. doi: 10.1007/BF02573070. [DOI] [PubMed] [Google Scholar]
  • 36.Bloch W, Huggel K, Sasaki T, et al. The angiogenesis inhibitor endostatin impairs blood vessel maturation during wound healing. FASEB J. 2000;14:2373–2376. doi: 10.1096/fj.00-0490fje. [DOI] [PubMed] [Google Scholar]
  • 37.Tanaka H, Taniguchi H, Mugitani T, et al. Angiogenesis inhibitor TNP-470 prevents implanted liver metastases after partial hepatectomy in an experimental model without impairing wound healing. Br J Surg. 1996;83:1444–1447. doi: 10.1002/bjs.1800831038. [DOI] [PubMed] [Google Scholar]
  • 38.Lange-Asschenfeldt B, Velasco P, Streit M, et al. The angiogenesis inhibitor vasostatin does not impair wound healing at tumor-inhibiting doses. J Invest Dermatol. 2001;117:1036–1041. doi: 10.1046/j.0022-202x.2001.01519.x. [DOI] [PubMed] [Google Scholar]
  • 39.Berger AC, Feldman AL, Gnant MF, et al. The angiogenesis inhibitor, endostatin, does not affect murine cutaneous wound healing. J Surg Res. 2000;91:26–31. doi: 10.1006/jsre.2000.5890. [DOI] [PubMed] [Google Scholar]
  • 40.Saif MW, Elfiky A, Salem RR. Gastrointestinal perforation due to bevacizumab in colorectal cancer. Ann Surg Oncol. 2007;14:1860–1869. doi: 10.1245/s10434-006-9337-9. [DOI] [PubMed] [Google Scholar]
  • 41.Sugrue MMK, Hainsworth J, Badarinath S, et al. Risk factors for gastrointestinal perforations in patients with metastatic colorectal cancer receiving bevacizumab plus chemotherapy. J Clin Oncol. 2006;24:3535. [Google Scholar]
  • 42.Lecarpentier E, Ouaffi L, Mir O, et al. Bevacizumab-induced small bowel perforation in a patient with breast cancer without intraabdominal metastases. Invest New Drugs. 2011;29:1500–1503. doi: 10.1007/s10637-010-9505-4. [DOI] [PubMed] [Google Scholar]
  • 43.Han ES, Monk BJ. What is the risk of bowel perforation associated with bevacizumab therapy in ovarian cancer? Gynecol Oncol. 2007;105:3–6. doi: 10.1016/j.ygyno.2007.01.038. [DOI] [PubMed] [Google Scholar]
  • 44.Schellhaas E, Loddenkemper C, Schmittel A, et al. Bowel perforation in non-small cell lung cancer after bevacizumab therapy. Invest New Drugs. 2009;27:184–187. doi: 10.1007/s10637-008-9162-z. [DOI] [PubMed] [Google Scholar]
  • 45.Wang Y, Fei D, Vanderlaan M, et al. Biological activity of bevacizumab, a humanized anti-VEGF antibody in vitro. Angiogenesis. 2004;7:335–345. doi: 10.1007/s10456-004-8272-2. [DOI] [PubMed] [Google Scholar]
  • 46.Heinzerling JH, Huerta S. Bowel perforation from bevacizumab for the treatment of metastatic colon cancer: Incidence, etiology, and management. Curr Surg. 2006;63:334–337. doi: 10.1016/j.cursur.2006.06.002. [DOI] [PubMed] [Google Scholar]
  • 47.Francone TD, Saleem A, Read TA, et al. Ultimate fate of the leaking intestinal anastomosis: Does leak mean permanent stoma? J Gastrointest Surg. 2010;14:987–992. doi: 10.1007/s11605-010-1190-2. [DOI] [PubMed] [Google Scholar]
  • 48.Bruce J, Krukowski ZH, Al-Khairy G, et al. Systematic review of the definition and measurement of anastomotic leak after gastrointestinal surgery. Br J Surg. 2001;88:1157–1168. doi: 10.1046/j.0007-1323.2001.01829.x. [DOI] [PubMed] [Google Scholar]
  • 49.Platell C, Barwood N, Dorfmann G, et al. The incidence of anastomotic leaks in patients undergoing colorectal surgery. Colorectal Dis. 2007;9:71–79. doi: 10.1111/j.1463-1318.2006.01002.x. [DOI] [PubMed] [Google Scholar]
  • 50.Richardson DL, Mariani A, Cliby WA. Risk factors for anastomotic leak after recto-sigmoid resection for ovarian cancer. Gynecol Oncol. 2006;103:667–672. doi: 10.1016/j.ygyno.2006.05.003. [DOI] [PubMed] [Google Scholar]
  • 51.Vergote I, Trope CG, Amant F, et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N Engl J Med. 2010;363:943–953. doi: 10.1056/NEJMoa0908806. [DOI] [PubMed] [Google Scholar]
  • 52.Carter J, Durfee J. A case of bowel perforation after neoadjuvant chemotherapy for advanced epithelial ovarian cancer. Gynecol Oncol. 2007;107:586–589. doi: 10.1016/j.ygyno.2007.09.008. [DOI] [PubMed] [Google Scholar]
  • 53.Seewaldt VL, Cain JM, Goff BA, et al. A retrospective review of paclitaxel-associated gastrointestinal necrosis in patients with epithelial ovarian cancer. Gynecol Oncol. 1997;67:137–140. doi: 10.1006/gyno.1997.4842. [DOI] [PubMed] [Google Scholar]
  • 54.Sfakianos GP, Numnum TM, Halverson CB, et al. The risk of gastrointestinal perforation and/or fistula in patients with recurrent ovarian cancer receiving bevacizumab compared to standard chemotherapy: A retrospective cohort study. Gynecol Oncol. 2009;114:424–426. doi: 10.1016/j.ygyno.2009.05.031. [DOI] [PubMed] [Google Scholar]
  • 55.Rose PG, Piver MS. Intestinal perforation secondary to paclitaxel. Gynecol Oncol. 1995;57:270–272. doi: 10.1006/gyno.1995.1140. [DOI] [PubMed] [Google Scholar]
  • 56.Katz S, Schulman N, Levin L. Free perforation in Crohn's disease: A report of 33 cases and review of literature. Am J Gastroenterol. 1986;81:38–43. [PubMed] [Google Scholar]
  • 57.Greenstein AJ, Mann D, Sachar DB, et al. Free perforation in Crohn's disease: I. A survey of 99 cases. Am J Gastroenterol. 1985;80:682–689. [PubMed] [Google Scholar]
  • 58.Greenstein AJ, Sachar DB, Mann D, et al. Spontaneous free perforation and perforated abscess in 30 patients with Crohn's disease. Ann Surg. 1987;205:72–76. doi: 10.1097/00000658-198701000-00013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Cappell MS, Batke M. Mechanical obstruction of the small bowel and colon. Med Clin North Am. 2008;92:575–597. doi: 10.1016/j.mcna.2008.01.003. viii. [DOI] [PubMed] [Google Scholar]
  • 60.D'Amato G, Rocha Lima C, Mahany JJ, et al. Neutropenic enterocolitis (typhilitis) associated with docetaxel therapy in a patient with non-small-cell lung cancer: Case report and review of literature. Lung Cancer. 2004;44:381–390. doi: 10.1016/j.lungcan.2003.11.018. [DOI] [PubMed] [Google Scholar]
  • 61.Katsumata N, Yasuda M, Takahashi F, et al. Dose-dense paclitaxel once a week in combination with carboplatin every 3 weeks for advanced ovarian cancer: A phase 3, open-label, randomised controlled trial. Lancet. 2009;374:1331–1338. doi: 10.1016/S0140-6736(09)61157-0. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Protocol

Articles from Journal of Clinical Oncology are provided here courtesy of American Society of Clinical Oncology

RESOURCES