Abstract
Upper gastrointestinal malignancies generally have moderate to poor cure rates, even in the earliest stages, thereby implying that both local and systemic treatments have room for improvement. Therapeutic options are broadening, however, with the development of new immunotherapies and targeted agents, which can have synergistic effects with radiotherapy. Here we discuss the current state of combined modality therapy for upper gastrointestinal malignancies, specifically recent successes and setbacks in trials of radiation therapy with targeted therapies, vaccines, immunotherapies, and chemotherapies.
Introduction
Recent data estimate that out of 1,665,540 new cancers diagnosed in the United States in 2014, upper gastrointestinal (UGI) malignancies comprised 130,650 (7.8%), with approximately 36% pancreatic, 33% hepatobiliary, 17% gastric, and 14% esophageal.1 The estimated mortality thereof was 92,660, of which approximately 43% were pancreatic, 29% were hepatobiliary, 16% esophageal, and 12% gastric. Pancreatic cancer in particular is the fourth-leading cause of death among all malignancies for both men and women in the United States. Overall, UGI cancers are often advanced, if not metastatic, at time of diagnosis,2–4 which in turn increases morbidity and mortality.
The aggressiveness of UGI cancers has led researchers to evaluate multimodal approaches to treatment involving combinations of surgery, radiotherapy, and systemic agents. Other authors have discussed the synergy between radiation and systemic therapies, for instance the radiosensitizing properties of systemic therapies5 as well as the potential “abscopal effect” of immunotherapy wherein local radiation facilitates distal control.6 We present findings from recent studies on combined modality regimens for UGI malignancies and the ramifications thereof on treatment.
Targeted Therapies
Monoclonal Antibodies
Targeted therapies have been a mainstay in treating certain cancers because of their ability to selectively target tumor cells while sparing normal cells. Monoclonal antibodies (MABs) are a category of targeted drugs that generally target specific proteins in growth factor and angiogenic pathways, which are often overactive or otherwise dysfunctional in tumor cells. MABs have been evaluated in combination with chemotherapy or with radiation therapy7 because of evidence that MABs can enhance radiation-induced apoptosis8 and promote cell cycle arrest,9 thereby preventing tumor repopulation. Recent studies aimed at using MABs with radiation therapy have shown promising results in treating UGI malignancies (Table 1).
Table 1.
Trials Combining Radiotherapy With Monoclonal Antibodies (MAB)
| Primary Investigators | Site (#Accrued) | Regimen | Agent and Target | Key Findings | Tolerability and Toxicity |
|---|---|---|---|---|---|
| Meng et al14 | Locally advanced esophageal SCC (55) | Paclitaxel, cisplatin, and 59.4 Gy (33 fractions) with cetuximab | Cetuximab, anti-EGFR MAB | 80% partial or complete response and 1 year OS 93%. | Grade 3 neutropenia and mucositis in 18/55 (33%) and 7/55 (13%), respectively. No Grade 4 toxicities observed. |
| Becerra et al16 | Esophageal ACA and GEJ (39) | Neoadjuvant cetuxiumab and 50.4 Gy (28 fractions) | Cetuximab, anti-EGFR MAB | 48% complete response rate 3 months after surgical resection. | Grade 5 aspiration seen in 1/39 (2%). Grade 3 dysphagia in 7/39 (17%). Grade 4 toxicities did not occur in 5% or more of patients. |
| Lee et al18 | Locally advanced esophageal ACA (19) | Neoadjuvant irinotecan, cisplatin, and cetuximab with 50.4 Gy (28 fractions) | Cetuximab, anti-EGFR MAB | Median OS 31 months and median PFS 10 months with 16% CR following surgery. | Grade 3–4 neutrophilia and dysphagia in 9/19 (47%) and 6/19 (31%), respectively. Grade 3 diarrhea also in 9/19 (47%). |
| Tomblyn et19 | Esophageal ACA (21) | Neoadjuvant irinotecan, cisplatin, and cetuximab with 50.4 Gy (28 fractions) | Cetuximba, anti-EGFR MAB | 2-years OS 33% and PFS 24% with overall response rate 18%. | Grade 3–4 toxicity in 76% (16/21), including 11/21 (52%) from hematologic derangements. GI necrosis and sudden death each occurred in 1/21 (5%). |
| Crosby et al21 | Esophageal ACA and SCC (258) | Cisplatin, capecitabine, and 50.4 Gy (28 fractions) with and without cetuximab | Cetuximab, anti-EGFR MAB | Cetuximab group had higher rate of treatment failure at 6 months (77%) and lower median OS (22 months). | Cetuximab group had more (102/129; 79%) nonhematological Grade 3–4 toxicities (eg, dysphagia, rash) vs control group (81/129; 63%). Control group had more (36/129; 28%) Grade 3–4 hematological toxicity vs cetuximab group (27/129; 21%). |
| Ilson et al22 | Esophageal ACA and SCC (328) | Capecitabine, paclitaxel, and 50.4 Gy (28 fractions) with and without cetuximab | Cetuximab, anti-EGFR MAB | No difference between cetuximab and control groups in terms of efficacy or safety. | Nearly equivalent rates of Grade 3 (45%−49%), 4 (17%−22%), and 5 (1%−4%) toxicities between groups. |
| Esnaola et al23 | Unresectable pancreatic ACA (37) | Gemcitabine, oxaliplatin, cetuximab, capecitabine, and 54 Gy (30 fractions) | Cetuximab, anti-EGFR MAB | Median OS and PFS of 12 months and 10 months, respectively. PFS after 6 months of 62%. No survival benefit seen | Grade 3–4 leukopenia in 5/37 (13%). Overall, well-tolerated regimen. |
| Zhao et al25 | Esophageal SCC (11) | Cisplatin, 5-FU, and 60.2 Gy (34 fractions) with nimotuzumab | Nimotuzumab, anti-EGFR MAB | 6-month OS 78%. 1-year OS and PFS of 67% and 100%, respectively. | No dose-limiting toxicity observed. Grade 3–4 esophagitis and leukopenia each seen in 2/11 (18%). |
| Ramos-Suzarte et al26 | Esophageal ACA and SCC (63) | Cisplatin, 5-FU, and 45–50 Gy (25–28 fractions) with and without nimotuzumab | Nimotuzumab, anti-EGFR MAB | Nimotuzumab group had better median OS and response rates. | Grade 3–4 diarrhea more common in nimotuzumab group (6/33; 18%) than in control (2/30; 7%). Well-tolerated regimen. |
| Lockhart et al96 | Locally advanced esophageal ACA (70) | Neoadjuvant docetaxel, cisplatin, panitumumab, and 50.4 Gy (28 fractions) | Panitumumab, anti-EGFR MAB | pCR rate 33% and near-pCR rate (ie, 10% or fewere viable cancer cells) another 20%. | Grade 3–4 esophagitis and lymphopenia seen in 13/70 (18%) and 30/70 (43%), respectively. Overall, Grade 4 toxicity in 48% (34/70) of patients. |
| Kordes et al97 | Locally advanced esophageal SCC and ACA (90) | Neoadjuvant carboplatin, paclitaxel, panitumumab, and 41.4 Gy (23 fractions) | Panitumumab, anti-EGFR MAB | Low CR rate of 22% overall with 14% in ACA and 47% with SCC. | Grade 3–4 rash and neutropenia in 10/90 (11%) and 9/90 (10%), respectively. |
| Bendell et al38 | Esophageal and GEJ ACA (62) | Neoadjuvant paclitaxel, carboplatin, 5-FU, and 45 Gy (25 fractions) with bevacizumab and erlotinib | Bevacizumab, anti-VEGF MAB Erlotinib, anti-EGFR TKI | CR = 29% with median OS and PFS of 30 months and 29 months, respectively. | Grade 3–4 leukopenia (40/62; 64%) and mucositis (26/62; 42%) were most common adverse effects. Grade 3–4 diarrhea and esophagitis each seen in 17/62 (27%). |
| Crane et al39 | Pancreatic ACA (82) | Capecitabine, bevacizumab, and 50.4 Gy (28 fractions) | Bevacizumab, anti-VEGF MAB | No survival benefit over standard therapy. | Overall, 29/82 (35%) had Grade 3 or higher GI toxicity. |
| Small et al40 | Pancreatic ACA (32) | Gemcitabine, bevacizumab, and 36 Gy (15 fractions) | Bevacizumab, anti-VEGF MAB | Improved PFS, but no survival benefit over standard therapy. | Grade 3 leukopenia (6/32; 19%) and nausea (5/32; 16%) were most common adverse events. |
Cetuximab
Cetuximab is an epidermal growth factor receptor (EGFR) inhibitor that has been used in colorectal cancer,10 as well as in squamous cell carcinomas of the head and neck.11 Recently, researchers have investigated its applicability in esophageal cancers, particularly since EGFR is widely expressed therein12 and generally portends a poor outcome.8,13 Of note, most of the studies involving EGFR therapies did not explicitly state whether EGFR status had been evaluated before enrollment.
Meng et al14 conducted a Phase II trial that combined cetuximab with paclitaxel, cisplatin, and 59.4 Gy delivered in 33 fractions to 55 patients with unresectable, locally advanced squamous cell carcinoma (SCC) of the esophagus. They found that 44/55 (88%) had a clinical response, including 29/55 (53%) with complete response and another 15/55 (27%) with partial response. The 1-year progression-free survival (PFS) and overall survival (OS) for the 45 patients who responded were 84.2% and 93.3%, respectively; at 2 years they were 74.9% and 80.0%, respectively. Their efficacy and safety outcomes were favorable (Table 1) and their trial was notable in that it included only patients with SCC histology, whereas other studies have also included adenocarcinoma (ACA) types.15
A Phase II trial of 39 patients (78% ACA and 37% gastroesophageal junction (GEJ) cancers) involved 2 weeks of cetuximab followed by neoadjuvant cetuximab with concurrent 50.4 Gy in 28 fractions, which in turn preceded surgical resection by 6–8 weeks.16 The pathological complete response (pCR) rate was 36.6% by intent-to-treat and 48% at 3 months in those patients who had an esophagectomy. Although there was no statistically significant difference in pCR between esophageal and GEJ sites, only 9/32 (28%) ACA patients achieved pCR whereas 6/9 (67%) SCC patients did, thereby suggesting that histology may predict response. pCR was also higher for those with IIA disease (70%) compared to those with IIB and III (28%–29%) disease. Toxicities and efficacy were comparable to what others found when evaluating neoadjuvant chemotherapy and chemoradiation in esophageal cancer.17
There have been, however, at least 2 studies that included cetuximab with preoperative irinotecan, cisplatin, and 50.4 Gy (28 fractions) for locally advanced esophageal cancer that produced less favorable results (Table 1).18,19 Lee et al18 enrolled 19 patients with IIA-IVA esophageal and GEJ cancer, most with ACA histology (84%) and 95% proceeded to surgery. Median PFS was 10 months and median OS was 31 months. Only 3/19 (16%) patients had a pCR, in contrast to the 25%–40% complete response rate seen in other studies using standard neoadjuvant chemoradiation with surgery.20 Tomblyn et al19 used a similar treatment schedule on 21 enrolled patients—48% ACA—with unresectable disease and found 2-years OS = 33.3% and PFS = 23.8% with overall response rate 17.6%. This was, however, a poorly tolerated regimen (Table 1). Thus, adding cetuximab to irinotecan, cisplatin, and radiotherapy led to worse efficacy and toxicity.
The SCOPE-1 trial by Crosby et al21 investigated the addition of cetuximab to definitive cisplatin-capecitabine-50 Gy in patients with nonmetastatic esophageal cancer. This was a multicenter Phase II-III randomized control trial wherein 129 patients were assigned to receive cetuximab with standard cisplatin-capecitabine with 50 Gy (25 fractions) whereas another 129 received standard therapy alone. Within each arm, approximately 70%–75% of patients had SCC histology, 25% had ACA type, and 60% had stage III disease. The study was terminated early because the experimental group had more treatment failures at 6 months (33.6% vs 23.1%) and grade 3–4 toxic events (79% vs 63%, P = 0.004), plus shorter median OS (22.1 months vs 25.4 months). Moreover, fewer patients in the experimental arm were able to complete full or reduced doses of cisplatin (77% vs 90%), capecitabine (69% vs 90%), or radiotherapy (81% vs 92%) due to toxicity or worsened disease. As such, the authors did not recommend incorporating cetuximab into cisplatin-capecitabine-based chemoradiation for localized esophageal cancer.
The radiation therapy oncology group (RTOG)-0436 multicenter Phase III trial by Ilson et al22 likewise showed no benefit when cetuximab was added to concurrent chemoradiation in nonresectable esophageal cancer. A total of 328 patients (62% ACA and 38% SCC) were randomized to receive cisplatin-paclitaxel-50.4 Gy (28 fractions) with or without cetuximab with OS as primary endpoint. CR was equivalent between the groups with 56% for those who had received cetuximab and 59% for those in the control arm. Moreover, the 1- and 2-year OS for the cetuximab group were 64% and 44%, respectively, compared to 65% and 42%, respectively, for the control group (P = 0.70). These OS rates remained largely similar when stratified for histology. The authors concluded that adding cetuximab to chemoradiation improved neither OS nor CR, thereby calling into question its role in esophageal cancer treatment.
Cetuximab has also been studied in both gastric and pancreatic cancer, but as of this writing we did not come across recent studies that investigated its use with radiotherapy in gastric cancer. One recent Phase II trial by Esnaola et al23 evaluated the safety and efficacy of induction gemcitabine-oxaliplatin with cetuximab followed by chemoradiation with capecitabine and 50.4 Gy (30 fractions) in locally advanced, unresectable pancreatic cancer. In the 37 patients involved, 30% underwent surgical resection; median OS was 11.8 months and median PFS was 10.4 months with 6-month PFS at 62%. Grade 3–4 toxicity was seen in around 30% after induction chemotherapy and less than 10% after chemoradiation. Although the treatment was well tolerated, neither PFS nor OS was ultimately improved that prompted the authors to call for more prospective studies.
Nimotuzumab
Nimotuzumab is another anti-EGFR MAB that has been evaluated with chemoradiation in squamous cell carcinomas of the head and neck.24 Zhao et al25 carried out a Phase I study to assess the maximum tolerated dose (MTD) and efficacy of nimotuzumab with concurrent cisplatin, 5 fluorouracil (5-FU), and 61.2 Gy (34 fractions) in locally advanced esophageal SCC. This regimen was well tolerated (Table 1) with 1-year OS and local PFS of 67% and 100%, respectively, which compared favorably with standard treatment.20
Ramos-Suzarte et al26 conducted a Phase II trial wherein 30 patients with unresectable Stage III–IV esophageal cancer were randomized to receive cisplatin, 5-FU, and 45.0–50 Gy (25–28 fractions of 1.8 Gy) and another 33 were assigned to have the same regimen plus nimotuzumab. EGFR expression was confirmed in a subset of patients (5 from control group and 13 from nimotuzumab group). The nimotuzumab group had a higher objective response rate (47.8%–15.4%), better local control rate (60.9%–26.9%), longer median survival (8.1 months to 2.9 months) and similar toxicity (Table 1). Thus, the nimotuzumab group had improved efficacy and comparable safety to standard treatment.
Nimotuzumab has been tested in both gastric and pancreatic cancer, but we could not find recent trials that included radiotherapy in the experimental regimens.
Trastuzumab
In addition to its prevalence in breast cancer, Human Epidermal Growth Factor Receptor-2 (HER-2) has been found to be overexpressed in at least 20% of patients with GEJ and both esophageal and gastric ACA tumors.27,28 There is a mortality benefit when HER-2-positive patients with both gastric and GEJ ACA receive trastuzumab, an anti-HER-2 MAB, with concurrent cisplatin-based chemotherapy.29 Trastuzumab also has radiosensitizing effects in esophageal cancer cell lines,30 which further enhances its appeal in combined modality therapy. The utility of HER-2 targeting in pancreatic and hepatobiliary malignancies, however, is less promising; a recent review by Omar et al31 found that HER-2 amplification in pancreatic and biliary malignancies ranged from 2%–24% and only 5%–8%, respectively. Xian et al32 had previously noted that HER-2 was amplified in just 1/868 (0.11%) of their patients with primary hepatocellular carcinoma (HCC).
Trastuzumab has not been well studied in conjunction with radiotherapy for UGI malignancies. Safran et al33 initially carried out a Phase I–II study wherein 19 patients with HER-2 positive, nonmetastatic esophageal ACA received cisplatin, paclitaxel, and 50.4 Gy (28 fractions) with trastuzumab. None of the patients had undergone surgical resection before enrollment and only one had surgical resection during the trial. A total of 74% (14/19) of the patients had celiac, retroperitoneal, portal, or scalene lymphadenopathy at enrollment, yet the authors found that their study group had a 2-years OS of 50% and median survival of 24 months, which was similar to what they had previously found without trastuzumab and without a notable difference in toxicity.34
The authors conceded that their small sample size necessitated further studies to evaluate efficacy and safety, which then led them to carry out the ongoing NCT01196390 Phase III trial involving patients with HER-2–positive esophageal and GEJ ACA. At the time of this writing, they have registered 194 patients to receive neoadjuvant paclitaxel, carboplatin, and 50.4 Gy (28 fractions) with or without trastuzumab. Results from this trial are still pending.
We did not find recent trials investigating the utility of trastuzumab with radiation in hepatobiliary or pancreatic cancer, which may be due to the low amplification levels cited earlier.
Bevacizumab
Vascular endothelial growth factor (VEGF) levels have been associated with esophageal and gastric cancer aggressiveness and mortality.35,36 As such, MABs that target VEGF, such as bevacizumab, have been evaluated for efficacy in various malignancies, including metastatic gastric cancer.37
Bendell et al38 tested the efficacy of bevacizumab and erlotinib with standard neoadjuvant paclitaxel, carboplatin, 5-FU, and 45.0 Gy (25 fractions) in treating localized esophageal and GEJ cancer. In their Phase II trial, 62 patients with resectable disease (93.5% with ACA) underwent treatment; 29% had pCR and the median OS and PFS were 30.2 months and 28.6 months, respectively. VEGF expression was not mentioned in the inclusion criteria. Although overall efficacy and toxicity were similar to those seen in standard neoadjuvant chemoradiation (Table 1), the authors found no evidence of improved efficacy and so did not feel further study of the specified regimen in esophageal cancer was warranted.
At least 2 other studies have investigated the applicability of bevacizumab to chemoradiation in localized pancreatic cancer. Crane et al evaluated the efficacy and safety of bevacizumab with capecitabine and 50.4 Gy (28 fractions) in patients with locally advanced, nonmetastatic pancreatic cancer.39 Among the 82 patients, the median and 1-year survival rates were 11.9 months and 47%, respectively, whereas the median and 1-year PFS were 8.6 months and 30%, respectively. Although toxicity was modest (Table 1), there was no survival benefit observed, so further evaluation was not advised.
Small et al40 later assessed the response rate, survival, and toxicity of gemcitabine-bevacizumab with 36 Gy of radiation in 15 fractions in 32 patients with nonmetastatic pancreatic cancer. At 10 weeks into treatment, 66% (21/32) had stable disease whereas another 9% (3/32) had partial or complete response; the remaining 4/32 (12%) had progressive disease. Overall, the authors found median PFS to be 9.9 months with PFS at 6 months, 1 year, and 2 years to be 62%, 38%, and 13%, respectively; median OS was 11.8 months with OS at 6 months, 1 year, and 2 years at 86%, 46%, and 23%, respectively. The regimen was well tolerated (Table 1), but while median PFS and 1-year PFS were moderately improved from that seen by the Crane et al study, no survival benefit was seen.
Tyrosine Kinase Inhibitors
Tyrosine kinase inhibitors (TKIs), like MABs, are typically engineered to target growth factor and angiogenic pathways. They differ in that while MABs are designed to prevent extracellular ligand-receptor interactions and subsequent receptor activation, the TKIs target adenosine triphosphate (ATP) binding sites on the cytoplasmic side of the receptor protein and thereby block downstream cascades.41 Much like MABs, TKIs have been found to exhibit immunomodulatory properties9 with radiotherapy, so their utility in combined modality approaches is of significant interest (Table 2).
Table 2.
Trials Combining Radiotherapy With Tyrosine Kinase Inhibitors (TKI)
| Primary Investigators | Site (#Accrued) | Regimen | Agent and Target | Key Findings | Tolerability and Toxicity |
|---|---|---|---|---|---|
| Li et al44 | Esophageal SCC (24) | Cisplatin, paclitaxel, erlotinib, and 60 Gy (30 fractions) | Erlotinib, anti-EGFR TKI | Improved OS, local control, and toxicity profile compared to RTOG-8501 | No Grade 4 toxicities. Most common Grade 3 toxicities were esophagitis and leukopenia, which were seen in 5/24 (21%) and in 4/24 (17%) patients, respectively. |
| Iyer et al46 | Esophageal ACA and SCC (17) | Erlotinib with 50.4 Gy (28 fractions) | Erlotinib, anti-EGFR TKI | Well-tolerated regimen with 1-year OS 29% | Grade 3–4 toxicities occurred in 5/17 (29.4%) patients; similar to RTOG-8501 trial. |
| Herman et al47 | Pancreatic ACA (48) | Adjuvant capecitabine, gemcitabine, erlotinib, and 50.4 Gy (28 fractions) | Erlotinib, anti-EGFR TKI | OS 93% at 1 year and 51% at 2 years. Recurrence-free survival 87% at 1 year and 30% at 2 years. | Grade 3–4 toxicities, mostly neutropenia, occurred in 26/48 (54%) patients with 16/26 (61%) during chemoradiation and 10/26 (48%) after maintenance chemotherapy. Unplanned hospitalizations needed in 16/48 (33%). |
| Konski et al50 | Pancreatic ACA (26) | Gemcitabine, erlotinib, and 28.0–28.8 Gy (0.5–0.6 Gy bid) | Erlotinib, anti-EGFR TKI | MTD not reached; median survival = 9.1 months and stable to improved disease in 88% | Grade 3–4 leukopenia seen in 14/26 (54%) patients. One patient had Grade 5 toxicity (bowel perforation). |
| Rodriguez et al52 | Esophageal and GEJ SCC and ACA (80) | Neoadjuvant cisplatin, 5-FU, geftinib, and 30 Gy (1.5 Gy bid) | Geftinib, anti-EGFR TKI | Geftinib showed superior OS compared to historic controls, but no difference in local or distal control. Gefitinib poorly tolerated. | Gefitinib-treated patients experienced less Grade 3–4 mucositis and (2/80; 2.5%), neutropenic fever (4/80; 5.0%), and fewer unplanned hospitalizations (11/80; 13.7%) compared with historic controls (14%, 17%, 28%, respectively). |
| Olsen et al53 | Pancreatic ACA (11) | Paclitaxel, geftinib, and 50.4 Gy (28 fractions) | Geftinib, anti-EGFR TKI | Poor survival | No Grade 4 toxicities seen. Diarrhea occurred in 10/11 (91 %), of which 3/10 (30%) were Grade 3. Fatigue seen in 9/11 (82%), of which 3/9 (33%) were Grade 3. |
| Chen et al55 | Hepatocellular carcinoma (40) | Sorafenib with 50–60 Gy (fractions of 2.0–2.5 Gy) then sequential sorafenib | Sorafenib, anti-VEGF-R TKI | 2-year OS 32% and in-field local control 39%. Increased toxicity with sequential sorafenib. | Grade 3–5 hepatic toxicity seen in 9/40 (22%) during sequential sorafenib, but during concurrent sorafenib-XRT only 4/40 (10%) had Grade 3 hepatotoxicity. No Grade 4 toxicities with concurrent sorafenib. Grade 1–2 GI ulcers and rashes very common (>90%). |
| Chiorean et al58 | Pancreatic ACA (27) | Gemcitabine, sorafenib, and 50 Gy (25 fractions) | Sorafenib, anti-VEGF-R TKI | MTD not reached, but no OS benefit. | No dose-limiting toxicity observed. Grade 3–4 GI bleeding and ulcers each in 7/27 (26%). |
| Aparicio et al60 | Pancreatic ACA (12) | Gemcitabine, sorafenib, and 45 Gy (25 fractions) | Sorafenib, anti-VEGF-R TKI | MTD not reached. No survival benefit. | Grade 4 posterior leukoencephalopathy and Grade 3 thrombocytopenia and fatigue each seen in 1/12 (8%). No other Grade 3–4 toxicities observed. |
Erlotinib
Erlotinib is a TKI that reversibly interacts with the adenosine triphosphate binding site of EGFR and has been shown to be an effective radiation sensitizer.42 Its favorable safety profile in esophageal carcinoma patients undergoing chemoradiation with cisplatin and 5-FU has been previously described.43 A 2010 Phase II study by Li et al44 evaluated its efficacy and toxicity with concurrent paclitaxel, cisplatin, and 60 Gy (30 fractions) for locally advanced esophageal SCC. They enrolled 24 patients and administered 6 weeks’ of treatment; a total of 11 (46%) had complete response whereas another 11 (46%) had partial response. At 2 years, their OS was 70.1% with disease-free survival (DFS of 57.4% and locoregional control rate of 87.5%; both OS and locoregional control rates were improved from those seen in RTOG-8501.45 Although 29% of the RTOG-8501 patients developed grade 3–4 toxicities, only 16.7% in the later study had similar events (Table 2).
Iyer et al46 also had encouraging results for their Phase II trial that looked at 17 (94% ACA) esophageal cancer patients ineligible for standard chemoradiation and whom they treated with concurrent erlotinib and 50.4 Gy in 28 fractions. Surgery was not included in the treatment. The median OS was 7.3 months with 1-year OS of 29% whereas median PFS was 4.5 months. Toxicity was similar to that seen for the RTOG-8501 trial (Table 2).
Erlotinib has also been studied in pancreatic cancer treatment. Herman et al47 published a Phase II trial that involved dosing erlotinib with concurrent adjuvant capecitabine and 50.4 Gy (28 fractions) followed by a course of erlotinib-gemcitabine, all following resection of stages I-II pancreatic ductal ACA. Among the 48 patients enrolled, OS and recurrence-free survival at 1 year were 93% and 87%, respectively. At 2 years, OS and recurrence-free survival dropped to 51% and 30%, respectively. Median OS was 24.4 months and median recurrence-free survival was 15.6 months. Their median OS and RFS as well as toxicity rates (Table 2) were similar to what other authors had found.48 A Phase I dose-escalation study of capecitabine-erlotinib with concurrent 50.4 Gy further supported the safety and feasibility of capecitabine-erlotinib with radiotherapy in locally advanced pancreatic cancer, although both median OS and PFS were only 11–12 months and 7 months, respectively.49
In a 2014 Phase I study, Konski et al50 attempted to use low-dose ultra-fractionated radiation to induce chemosensitization to gemcitabine and erlotinib in locally advanced pancreatic cancer. Patients were assigned to receive 0.4 Gy twice daily to a total of 28.8 Gy, 0.5 Gy twice daily to a total of 28.0 Gy, or 0.6 Gy twice daily to a total of 28.8 Gy. Median survival was 9.1 months with 8/26 having partial response and 15/26 having stable disease; the treatment was generally well tolerated and MTD was not reached (Table 2). The authors therefore encouraged further investigation of low-dose ultra-fractionated radiation in pancreatic cancer.
Overall, erlotinib has shown promising safety and efficacy in combined modality trials for both esophageal and pancreatic cancer, although further investigation with larger studies is needed.
Gefitinib
Gefitinib has a mechanism of action similar to that of erlotinib in that it targets EGFR and prevents downstream activation of antiapoptotic and proliferative signaling cascades; it differs mainly in having a different molecular structure.51 A Phase II study based in the Cleveland Clinic examined the utility of neoadjuvant gefitinib, cisplatin-5-FU, and 30 Gy (1.5 Gy twice daily) followed by surgical resection and then adjuvant gefitinib with 30 Gy (1.5 Gy twice daily) in locally advanced esophageal and GEJ cancer.52 Among the 80 patients enrolled, the authors found superior results in terms of OS (42% vs 28%) compared with a historic control group of patients who had received the same regimen, but without gefitinib (Table 2). There was no statistically significant difference, however, regarding distant metastatic control (40% vs 32%) and locoregional control (76% vs 77%). Thus, the authors advocated further assessment in light of the improved survival rate, but they conceded that almost half the patients could not tolerate maintenance gefitinib, which they attributed to the aggressive multimodality approach.
A smaller Phase I study investigated the combination of gefitinib with paclitaxel and 50.4 Gy (28 fractions) in 11 patients with locally advanced pancreatic cancer.53 Mean survival was 9 months from time of enrollment (range from <1 to 36 months) and toxicity fairly common (Table 2). Given the poor survival outcomes, the gefitinib-paclitaxel and radiation regimen is unlikely to merit continued evaluation despite having been well tolerated.
Sorafenib
Sorafenib is a TKI that targets VEGF receptor (VEGF-R), plasma-derived growth factor receptor, and numerous kinases commonly used in HCC.54 A 2013 Phase II study by Chen et al55 looked at concurrent sorafenib with radiation (50–60 Gy in fractions of 2.0–2.5 Gy) followed by sequential sorafenib in patients with unresectable, nonmetastatic HCC. A total of 40 patients with underlying cirrhosis were enrolled; 22/40 (55%) achieved partial to complete response whereas the other 18/40 (45%) had stable or worsened disease within a month of completing treatment. A 2-years OS and in-field progression free survival were 32% and 39%, respectively. Median OS was 14.0 months (95% CI: 9.1–18.9 months) whereas median in-field progression free survival was 8.9 months (95% CI: 2.4–15.3 months). Over 90% of patients developed Grade 1–2 gastroduodenal ulcers and hand-foot-skin reactions and some developed Grade 3 hepatotoxicity as well (Table 2).
In comparison, the 2013 Phase I–II trial by Bujold et al56 wherein 102 patients with locally advanced HCC received 30–54 Gy (6 fractions) via stereotactic body radiation therapy (SBRT) (without sorafenib) resulted in median OS = 17.0 months (95% CI: 10.4–21.3 months) and median time to progression 6.0 months (95% CI: 3.4–6.4 months). Around 36% of patients in the SBRT study developed at least Grade 3 toxicity, most of which (54%) were Grade 3 liver function panel derangements or thrombocytopenia. Indeed, a recent article by Pollom et al57 noted that VEGF inhibition may predispose patients to poor intestinal healing, ischemia, and even thromboembolism following injury from SBRT. Thus, while Chen et al highlighted the efficacy of sorafenib with radiation, the safety requires further assessment.
Two Phase I studies later evaluated sorafenib with simultaneous gemcitabine and radiotherapy in unresectable pancreatic ACA. Chiorean et al58 administered 50 Gy (25 fractions) along with gemcitabine (600 mg/m2 weekly) and sorafenib to 27 patients; no dose-limiting toxicity was observed (Table 2) and median OS was 12.6 months with median PFS 10.6 months. Unfortunately, the OS was not improved from standard protocols and about half the patients stopped treatment early.59 Aparicio et al60 carried out a similar study on 12 patients with gemcitabine (300 mg/m2 weekly), 45 Gy (25 fractions), and sorafenib. No dose-limiting toxicity was observed and median OS was 10 months whereas median PFS was just 4.4 months (Table 2). Thus, no survival benefit has been established yet for sorafenib with concurrent radiation in treating pancreatic cancer.
Nanoparticles and Conjugates
Nanotechnology is a bourgeoning field in oncology since the ability of nanoparticles to accumulate preferentially within tumors and deliver chemotherapeutic agents in slow, controlled fashion can potentially augment existing therapies.61 Recent studies, detailed in this section, have looked into the feasibility of using such molecules in conjunction with radiotherapy, especially since they can extravasate through the leaky tumor vasculature62,63 and exert radiation sensitivity effects within treatment fields.7
TNFerade
TNFerade is engineered from replication-deficient adenovirus containing tumor necrosis factor alpha (TNF-α) DNA ligated to radiation-inducible Egr-1 gene promoter.64 TNF-α helps induce apoptosis, cytotoxic free radical production, and immune cell recruitment, all of which can facilitate tumor cell destruction. Still, TNF-α produces a shock-like syndrome when infused systemically or directly into tumors, hence its incorporation into a virus, which can then pass through highly permeable tumor vessels and exert cytotoxic effects selectively within tumor.
A 2012 Phase I dose-escalation study by Chang et al65 to evaluate the efficacy of intratumoral TNFerade with neoadjuvant cisplatin, 5-FU, and concurrent 45 Gy (25 fractions) in locally advanced esophageal cancer resulted in median OS = 47.8 months and median DFS = 26.4 months. A total of 24 patients were enrolled with 20/24 (83%) ACA, 4/24 SCC (17%), and 20/24 (83%) having GEJ lesions. Ultimately, 21 patients were evaluated for clinical response with 20/21 (95%) having undergone surgical resection within 4–10 weeks of completing radiation treatment and 1/21 (5%) having an autopsy. The authors observed complete response in 6/21 patients (29%; 2/6 with SCC, and 4/6 with ACA) with 1-year and 3-years DFS rates 63% and 38%, respectively. The 1-year, 3-years, and 5-years OS rates were 88%, 54%, and 41%, respectively. The regimen was generally well tolerated, although the highest dose of TNFerade was associated with thromboembolic events. The authors concluded that TNFerade delivered endoscopically with concurrent 5-FU-cisplatin chemoradiation is efficacious and generally safe, but does not appear to change survival outcomes.
Another Phase I-II dose-escalation study that evaluated intratumoral TNFerade with concurrent 5-FU and 50.4 Gy (28 fractions) in locally advanced pancreatic cancer underscored the efficacy of TNFerade, with median OS of 9.8 months and median DFS of 3.6 months, both similar to standard 5-FU chemoradiation.66 Dose-limiting toxicities of pancreatitis and cholangitis were seen (3/50) in the highest-dose group, but overall the treatment was well tolerated. A Phase III trial by Herman et al67 had more sobering results; patients with locally advanced pancreatic cancer were randomly assigned to have standard 5-FU with 50.4 Gy either with or without TNFerade. Both groups had equivalent median OS of 10.0 months as well as median PFS of about 7 months. They also had almost similar survival rates at 1 year (41% in TNFerade group vs 36.7%) and at 2 years (11.3% in TNFerade group vs 10.3%). Median DFS was also similar at 11.6 months for the TNFerade group and 10.8 months for control group. The authors deemed TNFerade safe with mostly grade 2–3 toxicity, but ineffective in prolonging survival in locally advanced pancreatic cancer.
Liposomal Cisplatin (Lipoplatin)
Lipoplatin is a liposomal formulation of cisplatin; it is engineered to allow the large cisplatin molecule to be taken up by tumor cells and thereby concentrate within neoplastic masses.68 A 2010 Phase I–II study by Koukourakis et al69 involved treating 11 patients with locally advanced gastric cancer with 5-FU, lipoplatin, and radiotherapy to assess efficacy and toxicity. Grade 2–4 toxicity was uncommon (3/11), but the short median follow-up of 9 months could have precluded seeing evidence of longer-term morbidity or adverse events. Local control at 1 year was 51% and 1-year OS was 70%. Overall, the treatment was well tolerated, albeit in a small treatment group, and the authors called for further testing of lipoplatin with chemoradiation for gastric cancer.
CO-101 Lipid
CO-101 is a lipid-drug conjugate of gemcitabine that is designed to allow the medication to enter cells independently of levels of human equilibrative nucleoside transporter-1 (hENT-1) that can be down-regulated by tumor cells.70 Poplin et al71 evaluated its efficacy in metastatic pancreatic cancer in a randomized Phase II trial aimed at determining whether or not CO-101 could outperform gemcitabine depending on hENT-1 expression. They enrolled 367 patients and found no difference in OS within the gemcitabine arm between patients with low and high hENT-1 expression. Furthermore, differences in OS and toxicity profiles between the gemcitabine and CO-101 arms were not statistically significant. The authors deduced that hENT-1 expression is unlikely to predict gemcitabine sensitivity and, in any case, that CO-101 is not superior to gemcitabine in stage IV pancreatic cancer.
Paclitaxel-Poliglumex
Paclitaxel-poliglumex (PPX) is a conjugate of paclitaxel with biodegradable poly-l-glutamate to render it water-soluble in circulation, to facilitate tumor cell uptake via hyper-permeable neoplastic vasculature, and to enhance its radiation sensitizing effects.72 In a 2012 Phase II trial at Brown University, 40 patients with esophageal cancer (37 with ACA and 3 with SCC) were given PPX, cisplatin, and 50.4 Gy concurrent radiation to elucidate safety and treatment response.73 Three patients refused surgery, including 2 with SCC histology, but the remaining 37/40 (92%) underwent resection. No grade 4 toxicity was observed and the authors reported a 32% (12/37) complete response rate, all of whom had ACA. Thus, the study suggested that PPX could be a valuable radiation sensitizer in esophageal cancer treatment and that larger prospective studies would need to be undertaken to elucidate safety and efficacy.
Immunotherapies
Immunotherapy, specifically the direct recruitment of the immune system to attack tumors, has become an area of intensive research in recent years. Its incorporation into combined modality trials with radiation is of particular interest given the inherent immunomodulatory properties of radiotherapy.6 Indeed, others have written about the “abscopal effect” whereby localized radiation can induce regression or control of distant metastases because tumor antigens released from dying neoplastic cells potentiate an antibody-like response by the immune system.74 Although numerous trials of immunotherapeutic agents in UGI cancers are underway or being planned,75 we report the latest results of combined modality approaches using radiation and immunotherapy below.
Clivatuzumab Tetratexan
Clivatuzumab tetratexan is an MAB conjugated to yttrium-90 (Y90-hPAM4) that targets mucin-1 glycoprotein (MUC-1), which is produced by over 85% of pancreatic ACAs and which is largely absent from normal pancreatic tissue.76 Pancreatic cancer cells have shown sensitivity to radioimmunotherapy (RAIT), particularly when combined with gemcitabine, hence the interest in clivatuzumab tetratexan.77 In a 2012 Phase I trial, Ocean et al78 tested gemcitabine along with a fractionated schedule of clivatuzumab tetratexan in 38 patients with advanced pancreatic cancer. Median OS was 7.7 months, although 13 patients who received a repeat treatment cycle had a median OS of 11.8 months. Around 16% (6/38) had partial responses and another 42% (16/38) had disease stabilization. A total of 74% (28/38) developed grade III–IV thrombocytopenia or neutropenia after 1 cycle; this rate rose to 100% (38/38) if the patient opted for a repeated cycle. The authors felt that patients could tolerate fractionated radioimmunotherapy together with gemcitabine and called for further studies to investigate the promising therapeutic and safety indices.
GM-CSF Vaccine
Granulocyte-macrophage colony stimulating factor (GM-CSF) is an immunostimulant,79 so vaccine cells engineered to secrete GM-CSF have been evaluated as a potential adjunct to pancreatic cancer treatment. Jaffee et al80 designed irradiated tumor cells to secrete GM-CSF and thereby facilitate tumor antigen presentation by recruited dendritic cells and coordination among CD4, CD8, and NK cells. Lutz et al81 conducted a single institution Phase II study wherein 60 patients with resected pancreatic ACA received adjuvant 5-FU chemoradiation with GM-CSF immunotherapy to evaluate safety and efficacy. GM-CSF was delivered via irradiated and transfected allogeneic whole cell tumor lines. They reported median survival of 24.8 months and median DFS of 17.3 months; 1-year OS and DFS were 85.0% and 67.4%, respectively. Their median and 1-year OS was not significantly different from those seen in a historical cohort of patients treated with adjuvant chemoradiation alone at their institution. No dose-limiting toxicity was observed and most adverse effects consisted of dermatitis-type reactions that usually resolved within a week. The authors concluded that adjuvant GM-CSF immunotherapy was safe and efficacious; they also noted that mesothelin-specific T-cell frequencies induced by immunotherapy may be associated with improved DFS, and called for broadening scope of treatment in a multicenter study.
Checkpoint Inhibitors
Immune checkpoint inhibitors are among the most prominent immunotherapeutic agents within the past few years. These molecules target important stimulatory and inhibitory pathways in immune cells to modulate their tumor detection and cytotoxic abilities. Recent agents that target programmed death receptor-1 (PD-1)82 and programmed death receptor ligand-1 (PD-L1)83 have shown demonstrated efficacy in melanoma and other malignancies. Several authors have discussed the prevalence of PD-1 and PD-L1 overexpression in gastric and GEJ ACA84 as well as their negative effect on prognosis, which underscores the importance of testing checkpoint inhibitors in UGI malignancies. Some researchers have found that the anti-PD-1 MAB pembrolizumab has efficacy without severe toxicity in advanced gastric ACA85 as well as in both esophageal SCC and ACA.86 Others have evaluated ipilimumab that binds the inhibitory T-cell protein CTLA-4, together with gemcitabine in pancreatic ACA and found significant Grade 3–4 hematological adverse events (69%).87
Still, while the systemic efficacy of checkpoint inhibitors has been explored, we have not come across publications on checkpoint inhibitors with concurrent radiation for UGI malignancies as of this writing.
Other Agents
Eniluracil
Eniluracil is an irreversible inhibitor of dihydropyrimidine dehydrogenase, which usually breaks down 5-FU. Its utility in treating locally advanced pancreatic and biliary cancers with concurrent neoadjuvant 5-FU and 45 Gy (28 fractions) was assessed in a Phase I study by Czito et al.88 The patients received an additional 5.4 Gy (3 fractions) to gross tumor volume following initial radiation treatment. Among the 13 patients enrolled, MTD was not reached, only 1 dose-limiting adverse event was observed, and 3/13 (23%) had a pCR or CR. The authors showed that neoadjuvant eniluracil-5-FU and radiation was well tolerated and feasible, although further studies are lacking.
Triapine
Triapine is a radiosensitizer and ribonucleotide reductase inhibitor that prevents conversion of ribonucleoside diphosphates to deoxyribonucleotides, which in turn are used for DNA synthesis.89 In a 2012 Phase I dose-escalation study (n = 12), Martin et al90 investigated 3 levels of triapine with 50.4 Gy in patients with locally advanced pancreatic cancer. MTD was not reached, 6/12 (50%) had stable disease, 2/12 (17%) had partial response, and 11/12 (92%) had no local disease progression. The median time to first documented progression was 6.1 months with median OS of 6.8 months. There were 5 grade 4 hematologic events (lymphopenia), but no grade 4 nonhematologic toxicities. Moreover, ribonucleotide reductase levels were not predictive of outcomes. The authors concluded that triapine is well tolerated and may be useful with chemoradiation in future studies.
Amifostine
Amifostine is a cytoprotective agent that selectively protects normal tissue and scavenges free radicals and other reactive metabolites.91 Its utility in intrahepatic cancer was evaluated in a 2012 Phase I study by Feng et al92 to determine whether or not radiation dosing could be increased without significantly worsening radiation-induced liver disease. The authors enrolled 23 patients with diffuse intrahepatic malignancy—both primary and metastatic—and treated them with whole liver radiation to maximum dose 40 Gy with concurrent amifostine. They found that amifostine conferred a modest radioprotective effect in patients, particularly in primary liver cancer, up to 38 Gy; they could not estimate MTD in patients with liver metastases because of a few patients who previously received alkylating chemotherapy. Though they stopped short of saying amifostine would provide substantial benefit for whole liver radiation, the authors suggested that it could potentially allow higher radiation dosing for focal treatment.
IFN-α2b
Interferon-alfa-2b (IFN-α2b) has demonstrated radiosensitizing effects and synergy with other chemotherapeutic agents.93 In a 2011 Phase II trial of adjuvant cisplatin, 5-FU, and IFN-α2b with 50.4 Gy in 84 patients with pancreatic cancer, Picozzi et al94 reported median OS and DFS of 25.4 months and 14.1 months, respectively. The OS at 18 months was 69% and it dropped to 59% at 2 years. Both median and 2-years survival rates compared favorably with those seen in other trials of chemotherapy or chemoradiation for pancreatic cancer.95 No long-term toxicity or treatment-related deaths were observed, but they stated that 80/84 (95%) developed grade 3–4 toxicity events, including neutropenia and nausea. Thus, despite the encouraging efficacy seen in the trial and the absence of irreversible long-term toxicity, the safety profile of IFN-α2b was of concern to the authors and they noted that future investigations should explore quality-of-life measures.
Ongoing Trials or Future Directions
In summary, there have been numerous recent studies on combined modality approaches involving radiotherapy in treating UGI malignancies, mostly in esophageal and pancreatic tumors, and results have been varied. Among the targeted therapies, nimotuzumab, trastuzumab, and erlotinib have shown promise in esophageal cancers and the last of these has had encouraging results in pancreatic ACA as well. Unfortunately, cetuximab, bevacizumab, panitumumab, and gefitinib have not shown survival benefits and can even have higher toxicity risks. Sorafenib has shown efficacy, albeit with poor safety in hepatic tumors. TNFerade has been shown to be safe, but without survival benefit, in both esophageal and pancreatic cancers. Lipoplatin, paclitaxel, and IFN-α2b likely require further evaluation to determine safety and efficacy. The other agents—clivatuzumab, GM-CSF vaccine, and the rest—have shown good results in hepatobiliary and pancreatic tumors, but larger studies are needed to determine utility.
A recurrent theme in the literature is the failure to accrue enough patients to draw results that can influence current practice. This may be a reflection of fragmentation in the oncology community regarding trials for combined modality regimens as well as of the sheer array of novel agents to test. Though we concede that multicenter trials are challenging to initiate and carry out, it does appear that the time has come for larger, more collaborative endeavors to test the safety and efficacy of combined modality approaches. Moreover, we encourage further exploration of treatments for hepatobiliary and gastric malignancies as these were relatively underrepresented among the studies we reviewed compared to pancreatic and esophageal cancers. Even esophageal cancers can produce different outcomes based on histology, but all too often studies have not successfully included comparable numbers of SCC and ACA patients, perhaps from slow accrual. It may be easier to investigate these populations separately and thereby establish more definitive results for future reference. We also expect that the future would bring new studies investigating radiotherapy in conjunction with immunotherapies and checkpoint inhibitors for UGI neoplasms given the exciting developments seen in melanoma and non–small cell lung cancer. These agents represent a largely unknown quantity in UGI cancers and should be tested with radiation. Finally, we urge researchers to test other modalities of delivering radiation—proton beams, brachytherapy, or hypofractionation—in conjunction with systemic agents and different localized treatments like thermoablation, as these may yet yield surprising effects.
There are newer combined modality trials for UGI cancers (Table 3) besides the aforementioned NCT01196390. For instance, there is NCT01342224 at New York University wherein human telomerase vaccine is administered with GM-CSF, tadalafil, and 50.4 Gy (28 fractions) in locally advanced pancreatic ACA. The recently approved NCT02530437 trial at M.D. Anderson Cancer Center would examine the Hedgehog inhibitor taladegib with paclitaxel, carboplatin, and 50.4 Gy (28 fractions) in localized esophageal ACA and GEJ tumors. Overall, there is much optimism in combined modality approaches and we look forward to the results of the latest investigations.
Table 3.
Ongoing Trials Involving Systemic Approaches With Radiotherapy
| Clinical Trial Identifier | Cancer Type | Regimen | Agent and Target | Outcomes of Interest |
|---|---|---|---|---|
| NCT01196390; ongoing, but no longer recruiting | HER-2 (+) esophageal ACA | Neoadjuvant paclitaxel, carboplatin, and 50.4 Gy (28 fractions)with or without trastuzumab | Trastuzumab, anti-HER-2/Neu MAB | Disease-free survival, pCR, and toxicity over 8 years. Estimated 591 people enrolled. |
| NCT02530437; not yet recruiting | Esophageal and GEJ ACA | Paclitaxel, carboplatin, and 50.4 Gy (28 fractions) with either concurrent or preceding taladegib | Taladegib, Hedgehog inhibitor | Toxicity and pCR over 10 weeks. Goal enrollment 66 patients. |
| NCT02375581; currently recruiting | Esophageal SCC and ACA | Concurrent icotinib with 50–60 Gy (25–30 fractions) | Icotinib, anti-EGFR TKI | Mortality and toxicity over 2 years. Goal enrollment 130 patients. |
| NCT02545751; not yet recruiting | Metastatic esophageal SCC and ACA | Concurrent thymalfasin with 25 Gy (5 fractions) | Thymalfasin, stimulates immune system proliferation and differentiation | Tumor response over 8 weeks and toxicity and OS over 2 years. Goal enrollment 29 patients. |
| NCT02381561; not yet recruiting | Advanced UGI and lower GI cancers | Ropidoxuridine with concurrent radiotherapy (dosing dependent on cancer site) | Ropidoxuridine, prodrug for nucleoside analog idoxuridine | MTD over 28 days. Goal enrollment 30 patients. |
| NCT02425605; currently recruiting | Advanced HCC | Intra-arterial 5-FU with 45 Gy (25 fractions) followed by sorafenib | Sorafenib, anti-VEGF-R TKI | OS, PFS, and pCR over 36 months. Goal enrollment 47 patients. |
| NCT01342224; active, but no longer recruiting | Locally advanced pancreatic ACA | Neoadjuvant telomerase vaccine (GV1001), followed by GM-CSF with gemcitabine and 50.4 Gy (28 fractions). Tadalaf l given throughout treatment and after surgery. | Telomerase vaccine (GV1001), peptide subunit of telomerase reverse transcriptase that can generate T-cell response to telomerase Tadalafl, phosphodiesterase inhibitor | Toxicity and tumor response over 180 days. 11 enrolled. |
| NCT02439593; not yet recruiting | Locally advanced pancreatic ACA | Neoadjuvant FOLFIRINOX followed by concurrent gemcitabine and 50.4 Gy (28 fractions) with or without weekly hyperthermia (40–41C) | Localized hyperthermia to 40–41C | 1-year OS and safety. Goal 39 patients in each arm. |
| NCT02405585; currently recruiting | Borderline resectable pancreatic ACA | Neoadjuvant FOLFIRINOX followed by concurrent gemcitabine and 50.4 Gy (28 fractions) with monthly algenpantucel-l immunotherapy | Algenpantucel-L, immunotherapy with human pancreatic cancer cells that contain mouse gene, thereby inducing immune system to recognize and attack cancer cells as foreign | 18-month OS, PFS, CR, and safety. Goal 48 patients total. |
| NCT02349867; currently recruiting | Untreated pancreatic ACA | Neoadjuvant gemcitabine and nab-paclitaxel followed by concurrent gemcitabine, vorinostat, sorafenib, and 50.4 Gy (28 fractions) | Vorinostat, histone deacetylase inhibitor Sorafenib, anti-VEGF-R TKI | Determine optimal dose and schedule of vorinostat and sorafenib over 18–36 months. Goal 36 patients total. |
Footnotes
Conflicts of Interest: None.
References
- 1.Siegel R, Ma J, Zou Z, et al. : Cancer statistics, 2014. CA Cancer J Clin 64:9–29, 2014 [DOI] [PubMed] [Google Scholar]
- 2.Augustine MM, Fong Y: Epidemiology and risk factors of biliary tract and primary liver tumors. Surg Oncol Clin N Am 23:171–188, 2014 [DOI] [PubMed] [Google Scholar]
- 3.Rubenstein JH, Shaheen NJ: Epidemiology, diagnosis, and management of esophageal adenocarcinoma. Gastroenterology 149:[302.e301–317], 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Shah MA: Update on metastatic gastric and esophageal cancers. J Clin Oncol 33:1760–1769, 2015 [DOI] [PubMed] [Google Scholar]
- 5.Lawrence TS, Haffty BG, Harris JR: Milestones in the use of combined-modality radiation therapy and chemotherapy. J Clin Oncol 32:1173–1179, 2014 [DOI] [PubMed] [Google Scholar]
- 6.Tang C, Wang X, Soh H, et al. : Combining radiation and immunotherapy: A new systemic therapy for solid tumors? Cancer Immunol Res 2:831–838, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Morris ZS, Harari PM: Interaction of radiation therapy with molecular targeted agents. J Clin Oncol 32:2886–2893, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Milas L, Fan Z, Andratschke NH, et al. : Epidermal growth factor receptor and tumor response to radiation: In vivo preclinical studies. Int J Radiat Oncol Biol Phys 58:966–971, 2004 [DOI] [PubMed] [Google Scholar]
- 9.Scaringi C, Enrici RM, Minniti G: Combining molecular targeted agents with radiation therapy for malignant gliomas. Onco Targets Ther 6:1079–1095, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hubbard JM, Grothey A: Colorectal cancer in 2014: Progress in defining first-line and maintenance therapies. Nat Rev Clin Oncol 12:73–74, 2015 [DOI] [PubMed] [Google Scholar]
- 11.Bonner JA, Harari PM, Giralt J, et al. : Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 354:567–578, 2006 [DOI] [PubMed] [Google Scholar]
- 12.Takaoka M, Harada H, Andl CD, et al. : Epidermal growth factor receptor regulates aberrant expression of insulin-like growth factor-binding protein 3. Cancer Res 64:7711–7723, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gibson MK, Abraham SC, Wu TT, et al. : Epidermal growth factor receptor, p53 mutation, and pathological response predict survival in patients with locally advanced esophageal cancer treated with preoperative chemoradiotherapy. Clin Cancer Res 9:6461–6468, 2003 [PubMed] [Google Scholar]
- 14.Meng X, Wang J, Sun X, et al. : Cetuximab in combination with chemoradiotherapy in chinese patients with non-resectable, locally advanced esophageal squamous cell carcinoma: A prospective, multicenter phase II trail. Radiother Oncol 109:275–280, 2013 [DOI] [PubMed] [Google Scholar]
- 15.Safran H, Suntharalingam M, Dipetrillo T, et al. : Cetuximab with concurrent chemoradiation for esophagogastric cancer: Assessment of toxicity. Int J Radiat Oncol Biol Phys 70:391–395, 2008 [DOI] [PubMed] [Google Scholar]
- 16.Becerra CR, Hanna N, Mccollum AD, et al. : A phase II study with cetuximab and radiation therapy for patients with surgically resectable esophageal and ge junction carcinomas: Hoosier oncology group g05–92. J Thorac Oncol 8:1425–1429, 2013 [DOI] [PubMed] [Google Scholar]
- 17.Tepper J, Krasna MJ, Niedzwiecki D, et al. : Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol 26:1086–1092, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Lee MS, Mamon HJ, Hong TS, et al. : Preoperative cetuximab, irinotecan, cisplatin, and radiation therapy for patients with locally advanced esophageal cancer. Oncologist 18:281–287, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Tomblyn MB, Goldman BH, Thomas CR Jr., et al. : Cetuximab plus cisplatin, irinotecan, and thoracic radiotherapy as definitive treatment for locally advanced, unresectable esophageal cancer: A phase-II study of the swog (s0414). J Thorac Oncol 7:906–912, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Sjoquist KM, Burmeister BH, Smithers BM, et al. : Survival after neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal carcinoma: An updated meta-analysis. Lancet Oncol 12:681–692, 2011 [DOI] [PubMed] [Google Scholar]
- 21.Crosby T, Hurt CN, Falk S, et al. : Chemoradiotherapy with or without cetuximab in patients with oesophageal cancer (scope1): A multicentre, phase 2/3 randomised trial. Lancet Oncol 14:627–637, 2013 [DOI] [PubMed] [Google Scholar]
- 22.Ilson DH M J, Suntharalingam M, Dicker A, et al. : RTOG 0436: A phase III trial evaluating the addition of cetuximab to paclitaxel, cisplatin, and radiation for patients with esophageal cancer treated without surgery. J Clin Oncol 32(suppl): [abstract 4007], 2014 [Google Scholar]
- 23.Esnaola NF, Chaudhary UB, O’brien P, et al. : Phase 2 trial of induction gemcitabine, oxaliplatin, and cetuximab followed by selective capecitabine-based chemoradiation in patients with borderline resectable or unresectable locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys 88:837–844, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bonner JA, Harari PM, Giralt J, et al. : Radiotherapy plus cetuximab for locoregionally advanced head and neck cancer: 5-Year survival data from a phase 3 randomised trial, and relation between cetuximab-induced rash and survival. Lancet Oncol 11:21–28, 2010 [DOI] [PubMed] [Google Scholar]
- 25.Zhao KL, Hu XC, Wu XH, et al. : A phase I dose escalation study of nimotuzumab in combination with concurrent chemoradiation for patients with locally advanced squamous cell carcinoma of esophagus. Invest New Drugs 30:1585–1590, 2012 [DOI] [PubMed] [Google Scholar]
- 26.Ramos-Suzarte M, Lorenzo-Luaces P, Lazo NG, et al. : Treatment of malignant, non-resectable, epithelial origin esophageal tumours with the humanized anti-epidermal growth factor antibody nimotuzumab combined with radiation therapy and chemotherapy. Cancer Biol Ther 13:600–605, 2012 [DOI] [PubMed] [Google Scholar]
- 27.Janjigian YY, Werner D, Pauligk C, et al. : Prognosis of metastatic gastric and gastroesophageal junction cancer by HER2 status: A European and USA international collaborative analysis. Ann Oncol 23:2656–2662, 2012 [DOI] [PubMed] [Google Scholar]
- 28.Nahta R, Hortobágyi GN, Esteva FJ: Growth factor receptors in breast cancer: Potential for therapeutic intervention. The Oncologist 8:5–17, 2003 [DOI] [PubMed] [Google Scholar]
- 29.Bang YJ, Van Cutsem E, Feyereislova A, et al. : Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (TOGA): A phase 3, open-label, randomised controlled trial. Lancet 376:687–697, 2010 [DOI] [PubMed] [Google Scholar]
- 30.Sato S, Kajiyama Y, Sugano M, et al. : Monoclonal antibody to HER-2/neu receptor enhances radiosensitivity of esophageal cancer cell lines expressing HER-2/neu oncoprotein. Int J Radiat Oncol Biol Phys 61:203–211, 2005 [DOI] [PubMed] [Google Scholar]
- 31.Omar N, Yan B, Salto-Tellez M: HER2: An emerging biomarker in non-breast and non-gastric cancers. Pathogenesis 2:1–9, 2015 [Google Scholar]
- 32.Xian ZH, Zhang SH, Cong WM, et al. : Overexpression/amplification of HER-2/neu is uncommon in hepatocellular carcinoma. J Clin Pathol 58:500–503, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Safran H, Dipetrillo T, Akerman P, et al. : Phase I/II study of trastuzumab, paclitaxel, cisplatin and radiation for locally advanced, HER2 over-expressing, esophageal adenocarcinoma. Int J Radiat Oncol Biol Phys 67:405–409, 2007 [DOI] [PubMed] [Google Scholar]
- 34.Safran H, Gaissert H, Akerman P, et al. : Paclitaxel, cisplatin, and concurrent radiation for esophageal cancer. Cancer Invest 19:1–7, 2001 [DOI] [PubMed] [Google Scholar]
- 35.Juttner S, Wissmann C, Jons T, et al. : Vascular endothelial growth factor-D and its receptor VEGFR-3: Two novel independent prognostic markers in gastric adenocarcinoma. J Clin Oncol 24:228–240, 2006 [DOI] [PubMed] [Google Scholar]
- 36.Rades D, Golke H, Schild SE, et al. : Impact of VEGF and VEGF receptor 1 (FLT1) expression on the prognosis of stage III esophageal cancer patients after radiochemotherapy. Strahlenther Onkol 184:416–420, 2008 [DOI] [PubMed] [Google Scholar]
- 37.Ohtsu A, Shah MA, Van Cutsem E, et al. : Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: A randomized, double-blind, placebo-controlled phase III study. J Clin Oncol 29:3968–3976, 2011 [DOI] [PubMed] [Google Scholar]
- 38.Bendell JC, Meluch A, Peyton J, et al. : A phase II trial of preoperative concurrent chemotherapy/radiation therapy plus bevacizumab/erlotinib in the treatment of localized esophageal cancer. Clin Adv Hematol Oncol 10:430–437, 2012 [PubMed] [Google Scholar]
- 39.Crane CH, Winter K, Regine WF, et al. : Phase II study of bevacizumab with concurrent capecitabine and radiation followed by maintenance gemcitabine and bevacizumab for locally advanced pancreatic cancer: Radiation therapy oncology group RTOG 0411. J Clin Oncol 27:4096–4102, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Small W Jr, Mulcahy MF, Rademaker A, et al. : Phase II trial of full-dose gemcitabine and bevacizumab in combination with attenuated three-dimensional conformal radiotherapy in patients with localized pancreatic cancer. Int J Radiat Oncol Biol Phys 80:476–482, 2011 [DOI] [PubMed] [Google Scholar]
- 41.Nyati MK, Morgan MA, Feng FY, et al. : Integration of EGFR inhibitors with radiochemotherapy. Nat Rev Cancer 6:876–885, 2006 [DOI] [PubMed] [Google Scholar]
- 42.Chinnaiyan P, Huang S, Vallabhaneni G, et al. : Mechanisms of enhanced radiation response following epidermal growth factor receptor signaling inhibition by erlotinib (tarceva). Cancer Res 65:3328–3335, 2005 [DOI] [PubMed] [Google Scholar]
- 43.Dobelbower MC, Russo SM, Raisch KP, et al. : Epidermal growth factor receptor tyrosine kinase inhibitor, erlotinib, and concurrent 5-fluorouracil, cisplatin and radiotherapy for patients with esophageal cancer: A phase I study. Anticancer Drugs 17:95–102, 2006 [DOI] [PubMed] [Google Scholar]
- 44.Li G, Hu W, Wang J, et al. : Phase II study of concurrent chemoradiation in combination with erlotinib for locally advanced esophageal carcinoma. Int J Radiat Oncol Biol Phys 78:1407–1412, 2010 [DOI] [PubMed] [Google Scholar]
- 45.Cooper JS, Guo MD, Herskovic A, et al. : Chemoradiotherapy of locally advanced esophageal cancer: Long-term follow-up of a prospective randomized trial (RTOG 85–01). Radiation therapy oncology group. J Am Med Assoc 281:1623–1627, 1999 [DOI] [PubMed] [Google Scholar]
- 46.Iyer R, Chhatrala R, Shefter T, et al. : Erlotinib and radiation therapy for elderly patients with esophageal cancer—Clinical and correlative results from a prospective multicenter phase 2 trial. Oncology 85:53–58, 2013 [DOI] [PubMed] [Google Scholar]
- 47.Herman JM, Fan KY, Wild AT, et al. : Phase 2 study of erlotinib combined with adjuvant chemoradiation and chemotherapy in patients with resectable pancreatic cancer. Int J Radiat Oncol Biol Phys 86:678–685, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bao PQ, Ramanathan RK, Krasinkas A, et al. : Phase II study of gemcitabine and erlotinib as adjuvant therapy for patients with resected pancreatic cancer. Ann Surg Oncol 18:1122–1129, 2011 [DOI] [PubMed] [Google Scholar]
- 49.Jiang Y, Mackley HB, Kimchi ET, et al. : Phase I dose escalation study of capecitabine and erlotinib concurrent with radiation in locally advanced pancreatic cancer. Cancer Chemother Pharmacol 74:205–210, 2014 [DOI] [PubMed] [Google Scholar]
- 50.Konski A, Meyer JE, Joiner M, et al. : Multi-institutional phase I study of low-dose ultra-fractionated radiotherapy as a chemosensitizer for gemcitabine and erlotinib in patients with locally advanced or limited metastatic pancreatic cancer. Radiother Oncol 113:35–40, 2014 [DOI] [PubMed] [Google Scholar]
- 51.Bronte G, Rolfo C, Giovannetti E, et al. : Are erlotinib and gefitinib interchangeable, opposite or complementary for non-small cell lung cancer treatment? Biological, pharmacological and clinical aspects. Crit Rev Oncol Hematol 89:300–313, 2014 [DOI] [PubMed] [Google Scholar]
- 52.Rodriguez CP, Adelstein DJ, Rice TW, et al. : A phase II study of perioperative concurrent chemotherapy, gefitinib, and hyperfractionated radiation followed by maintenance gefitinib in locoregionally advanced esophagus and gastroesophageal junction cancer. J Thorac Oncol 5:229–235, 2010 [DOI] [PubMed] [Google Scholar]
- 53.Olsen CC, Schefter TE, Chen H, et al. : Results of a phase I trial of 12 patients with locally advanced pancreatic carcinoma combining gefitinib, paclitaxel, and 3-dimensional conformal radiation: Report of toxicity and evaluation of circulating K-ras as a potential biomarker of response to therapy. Am J Clin Oncol 32:115–121, 2009 [DOI] [PubMed] [Google Scholar]
- 54.Llovet JM, Ricci S, Mazzaferro V, et al. : Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 359:378–390, 2008 [DOI] [PubMed] [Google Scholar]
- 55.Chen SW, Lin LC, Kuo YC, et al. : Phase 2 study of combined sorafenib and radiation therapy in patients with advanced hepatocellular carcinoma. Int J Radiat Oncol Biol Phys 88:1041–1047, 2014 [DOI] [PubMed] [Google Scholar]
- 56.Bujold A, Massey CA, Kim JJ, et al. : Sequential phase I and II trials of stereotactic body radiotherapy for locally advanced hepatocellular carcinoma. J Clin Oncol 31:1631–1639, 2013 [DOI] [PubMed] [Google Scholar]
- 57.Pollom EL, Deng L, Pai RK, et al. : Gastrointestinal toxicities with combined antiangiogenic and stereotactic body radiation therapy. Int J Radiat Oncol Biol Phys 92:568–576, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chiorean EG, Schneider BP, Akisik FM, et al. : Phase 1 pharmacogenetic and pharmacodynamic study of sorafenib with concurrent radiation therapy and gemcitabine in locally advanced unresectable pancreatic cancer. Int J Radiat Oncol Biol Phys 89:284–291, 2014 [DOI] [PubMed] [Google Scholar]
- 59.Loehrer PJ Sr, Feng Y, Cardenes H, et al. : Gemcitabine alone versus gemcitabine plus radiotherapy in patients with locally advanced pancreatic cancer: An eastern cooperative oncology group trial. J Clin Oncol 29:4105–4112, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Aparicio J, Garcia-Mora C, Martin M, et al. : A phase I, dose-finding study of sorafenib in combination with gemcitabine and radiation therapy in patients with unresectable pancreatic adenocarcinoma: A grupo espanol multidisciplinario en cancer digestivo (gemcad) study. PLoS One 9: e82209, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wang AZ, Tepper JE: Nanotechnology in radiation oncology. J Clin Oncol 32:2879–2885, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Jain RK, Stylianopoulos T: Delivering nanomedicine to solid tumors. Nat Rev Clin Oncol 7:653–664, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wang AZ, Langer R, Farokhzad OC: Nanoparticle delivery of cancer drugs. Annu Rev Med 63:185–198, 2012 [DOI] [PubMed] [Google Scholar]
- 64.Gupta VK, Park JO, Jaskowiak NT, et al. : Combined gene therapy and ionizing radiation is a novel approach to treat human esophageal adenocarcinoma. Ann Surg Oncol 9:500–504, 2002 [DOI] [PubMed] [Google Scholar]
- 65.Chang KJ, Reid T, Senzer N, et al. : Phase I evaluation of TNFerade biologic plus chemoradiotherapy before esophagectomy for locally advanced resectable esophageal cancer. Gastrointest Endosc 75:[1139.e1132–1146], 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hecht JR, Farrell JJ, Senzer N, et al. : Eus or percutaneously guided intratumoral TNFerade biologic with 5-fluorouracil and radiotherapy for first-line treatment of locally advanced pancreatic cancer: A phase I/II study. Gastrointest Endosc 75:332–338, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Herman JM, Wild AT, Wang H, et al. : Randomized phase III multi-institutional study of tnferade biologic with fluorouracil and radiotherapy for locally advanced pancreatic cancer: Final results. J Clin Oncol 31:886–894, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Stathopoulos GP, Boulikas T, Vougiouka M, et al. : Pharmacokinetics and adverse reactions of a new liposomal cisplatin (lipoplatin): Phase I study. Oncol Rep 13:589–595, 2005 [PubMed] [Google Scholar]
- 69.Koukourakis MI, Giatromanolaki A, Pitiakoudis M, et al. : Concurrent liposomal cisplatin (lipoplatin), 5-fluorouracil and radiotherapy for the treatment of locally advanced gastric cancer: A phase I/II study. Int J Radiat Oncol Biol Phys 78:150–155, 2010 [DOI] [PubMed] [Google Scholar]
- 70.Farrell JJ, Elsaleh H, Garcia M, et al. : Human equilibrative nucleoside transporter 1 levels predict response to gemcitabine in patients with pancreatic cancer. Gastroenterology 136:187–195, 2009 [DOI] [PubMed] [Google Scholar]
- 71.Poplin E, Wasan H, Rolfe L, et al. : Randomized, multicenter, phase ii study of CO-101 versus gemcitabine in patients with metastatic pancreatic ductal adenocarcinoma: Including a prospective evaluation of the role of HENT1 in gemcitabine or CO-101 sensitivity. J Clin Oncol 31:4453–4461, 2013 [DOI] [PubMed] [Google Scholar]
- 72.Li C, Ke S, Wu QP, et al. : Tumor irradiation enhances the tumor-specific distribution of poly(l-glutamic acid)-conjugated paclitaxel and its anti-tumor efficacy. Clin Cancer Res 6:2829–2834, 2000 [PubMed] [Google Scholar]
- 73.Dipetrillo T, Suntharalingam M, Ng T, et al. : Neoadjuvant paclitaxel poliglumex, cisplatin, and radiation for esophageal cancer: A phase 2 trial. Am J Clin Oncol 35:64–67, 2012 [DOI] [PubMed] [Google Scholar]
- 74.Formenti SC, Demaria S: Combining radiotherapy and cancer immunotherapy: A paradigm shift. J Natl Cancer Inst 105:256–265, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Murphy A, Kelly RJ: Immunotherapy in upper gi malignancies. Curr Treat Options Oncol 16:20, 2015 [DOI] [PubMed] [Google Scholar]
- 76.Gold DV, Karanjawala Z, Modrak DE, et al. : PAM4-reactive MUC1 is a biomarker for early pancreatic adenocarcinoma. Clin Cancer Res 13:7380–7387, 2007 [DOI] [PubMed] [Google Scholar]
- 77.Gold DV, Modrak DE, Schutsky K, et al. : Combined 90yttrium-DOTA-labeled PAM4 antibody radioimmunotherapy and gemcitabine radio-sensitization for the treatment of a human pancreatic cancer xenograft. Int J Cancer 109:618–626, 2004 [DOI] [PubMed] [Google Scholar]
- 78.Ocean AJ, Pennington KL, Guarino MJ, et al. : Fractionated radioimmunotherapy with (90) y-clivatuzumab tetraxetan and low-dose gemcitabine is active in advanced pancreatic cancer: A phase 1 trial. Cancer 118:5497–5506, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Dillman RO, Wiemann M, Nayak SK, et al. : Interferon-gamma or granulocyte-macrophage colony-stimulating factor administered as adjuvants with a vaccine of irradiated autologous tumor cells from short-term cell line cultures: A randomized phase 2 trial of the cancer biotherapy research group. J Immunother 26:367–373, 2003 [DOI] [PubMed] [Google Scholar]
- 80.Jaffee EM, Hruban RH, Biedrzycki B, et al. : Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: A phase I trial of safety and immune activation. J Clin Oncol 19:145–156, 2001 [DOI] [PubMed] [Google Scholar]
- 81.Lutz E, Yeo CJ, Lillemoe KD, et al. : A lethally irradiated allogeneic granulocyte-macrophage colony stimulating factor-secreting tumor vaccine for pancreatic adenocarcinoma. A phase II trial of safety, efficacy, and immune activation. Ann Surg 253:328–335, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Topalian SL, Hodi FS, Brahmer JR, et al. : Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366:2443–2454, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Brahmer JR, Tykodi SS, Chow LQ, et al. : Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366:2455–2465, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Raufi AG, Klempner SJ: Immunotherapy for advanced gastric and esophageal cancer: Preclinical rationale and ongoing clinical investigations. J Gastrointest Oncol 6:561–569, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Bang YJ, Chung HC, Shankaran V, et al. : Relationship between PD-L1 expression and clinical outcomes in patients with advanced gastric cancer treated with the anti-PD-1 monoclonal antibody pembrolizumab (MK-3475) in KEYNOTE-012. J Clin Oncol 33(suppl):[abstract 4001], 2015 [Google Scholar]
- 86.Doi T, Piha-Paul SA, Jalal SI, et al. : Pembrolizumab (MK-3475) for patients (PTS) with advanced esophageal carcinoma: Preliminary results from KEYNOTE-028. J Clin Oncol 33(suppl):[abstract 4010], 2015 [Google Scholar]
- 87.Mohindra NA, Kircher SM, Nimeiri HS, et al. : Results of the phase Ib study of ipilimumab and gemcitabine for advanced pancreas cancer. J Clin Oncol 33(suppl):[abstract e15281], 2015 [Google Scholar]
- 88.Czito BG, Hong TJ, Cohen DP, et al. : A phase I study of eniluracil/5-FU in combination with radiation therapy for potentially resectable and/or unresectable cancer of the pancreas and distal biliary tract. Cancer Invest 24:9–17, 2006 [DOI] [PubMed] [Google Scholar]
- 89.Barker CA, Burgan WE, Carter DJ, et al. : In vitro and in vivo radio-sensitization induced by the ribonucleotide reductase inhibitor triapine (3-aminopyridine-2-carboxaldehyde-thiosemicarbazone). Clin Cancer Res 12:2912–2918, 2006 [DOI] [PubMed] [Google Scholar]
- 90.Martin LK, Grecula J, Jia G, et al. : A dose escalation and pharmacodynamic study of triapine and radiation in patients with locally advanced pancreas cancer. Int J Radiat Oncol Biol Phys 84:e475–481, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Peters GJ, Van Der Vijgh WJ: Protection of normal tissues from the cytotoxic effects of chemotherapy and radiation by amifostine (WR-2721): Preclinical aspects. Eur J Cancer 31A(suppl 1):S1–S7, 1995 [DOI] [PubMed] [Google Scholar]
- 92.Feng M, Smith DE, Normolle DP, et al. : A phase I clinical and pharmacology study using amifostine as a radioprotector in dose-escalated whole liver radiation therapy. Int J Radiat Oncol Biol Phys 83:1441–1447, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Holsti LR, Mattson K, Niiranen A, et al. : Enhancement of radiation effects by alpha interferon in the treatment of small cell carcinoma of the lung. Int J Radiat Oncol Biol Phys 13:1161–1166, 1987 [DOI] [PubMed] [Google Scholar]
- 94.Picozzi VJ, Abrams RA, Decker PA, et al. : Multicenter phase II trial of adjuvant therapy for resected pancreatic cancer using cisplatin, 5-fluorouracil, and interferon-alfa-2b-based chemoradiation: ACOSOG Trial Z05031. Ann Oncol 22:348–354, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Neoptolemos JP, Stocken DD, Friess H, et al. : A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med 350:1200–1210, 2004 [DOI] [PubMed] [Google Scholar]
- 96.Lockhart AC, Reed CE, Decker PA, et al. : Phase II study of neoadjuvant therapy with docetaxel, cisplatin, panitumumab, and radiation therapy followed by surgery in patients with locally advanced adenocarcinoma of the distal esophagus (ACOSOG Z4051). Ann Oncol 25:1039–1044, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kordes S, Van Berge Henegouwen MI, Hulshof MC, et al. : Preoperative chemoradiation therapy in combination with panitumumab for patients with resectable esophageal cancer: The pact study. Int J Radiat Oncol Biol Phys 90:190–196, 2014 [DOI] [PubMed] [Google Scholar]
