67.04 Survival in DCIS Patients Undergoing Surgery versus Patients Not Undergoing Surgery

P. Singh1, M. Miller2, C. Wang4, D. J. Winchester3, C. Pesce3, E. Barrera3, K. Yao3  1University Of Chicago,Surgery,Chicago, IL, USA 2Memorial Sloan-Kettering Cancer Center,Surgery,New York, NY, USA 3Northshore University Health System,Surgery,Evanston, IL, USA 4Northshore University Health System,Center For Biomedical Research Informatics,Evanston, IL, USA

Introduction:  A clinical trial in Europe is randomizing low-grade ductal carcinoma in situ (DCIS) patients to surgery versus no surgery arms and a similar trial will start soon in the United States. We utilized the National Cancer Database (NCDB) and the Surveillance, Epidemiology, and End Results (SEER) retrospective databases to examine overall (OS) and disease-specific (DSS) survival between patients who underwent surgical excision of DCIS versus those patients who did not undergo excision.

Methods:  The NCDB and SEER databases were queried for low-grade DCIS patients treated from 2004-2011. We examined OS from both databases and DSS from SEER alone and used Cox regression modeling to adjust for patient, tumor and treatment factors. Mean follow-up for the surgery group was 5.6 years and 5.1 years for the no surgery group.

Results: Of 150,479 low-grade DCIS patients from the NCDB, 2,470 (2.0%) were reported to have no surgery and of 17,342 low-grade DCIS patients from SEER, 443 (2.5%) had no surgery. The mean age of the surgery group for NCDB and SEER was 58.8 and 58.4 years respectively and for the no surgery group was 60.4 and 60.8 years respectively. Mean tumor size was 1.5cm and 1.6cm for the NCDB and SEER patients respectively who had surgery, and 1.8cm and 1.5cm respectively for those who did not have surgery. Estrogen receptor was positive in 82.5% and 83.3% of the NCDB and SEER patients who had surgery and 82.1% and 85.8% of the NCDB and SEER patients who did not have surgery. In the NCDB and SEER patients undergoing surgery, 48.3% and 52.8% had radiation therapy versus 16.7% and 7.9% of those not undergoing surgery. Of NCDB patients, 33.1% of the surgery group received hormonal therapy compared to 11.7% in the no surgery group. OS at 10 years for both NCDB and SEER patients was greater for those who had surgery versus those who did not; in SEER, DSS was greater in the surgery group (Table 1). On Cox regression modeling adjusting for patient, tumor and treatment factors, not having surgery was associated with a 2.07 (95%CI: 1.73-2.46, p<0.001) greater risk of death in the NCDB and a 2.05 (95%CI: 1.63-2.59, p<0.001) greater risk of death in the SEER patients. The hazard ratio for DSS was 6.37 (95%CI: 3.8-10.6, p<0.0001) in SEER. Similar findings were found for estrogen receptor-positive patients alone.

Conclusion: OS and DSS were significantly higher in the surgery group versus the no surgery group. Although findings were similar between the two databases, selection bias may account for the survival differences between the surgery and no surgery groups given the retrospective nature of the databases. Future clinical trials will be more definitive in determining survival outcomes in DCIS patients not undergoing surgery.

 

67.03 Patients with Low or High BMI are at Higher Risk for Nipple/Skin Loss After Nipple Sparing Mastectomy

E. M. Urrechaga1, A. Soran1,2, P. F. McAuliffe1,2,3, R. R. Johnson1,2, C. Thomas1,2, M. Bonaventura1,2, G. M. Ahrendt1,2, E. J. Diego1,2  3Magee-Womens Research Institute,Pittsburgh, PA, USA 1University Of Pittsburgh,Pittsburgh, PA, USA 2Magee-Womens Hospital Of UPMC,Pittsburgh, PA, USA

Introduction:

Nipple sparing mastectomy (NSM) has been proven to be oncologically safe with relatively low complication rates. With expanding indications for NSM, a larger population of patients (pts) is undergoing the procedure for therapeutic or prophylactic purposes.  Though it has been established that obesity (Body Mass Index (BMI) >30) increases the risk for complications after NSM requiring reoperation, there is a paucity of data regarding this risk in patients with a low BMI (<18.5). We hypothesize that patients with a low BMI are at higher risk of clinically significant nipple and skin complications requiring a reoperation after NSM compared to patients with a normal BMI.

Methods:  

A retrospective review of a prospectively maintained NSM registry was performed at a single institution from August 2010-June 2016. Clinicopathological information including age, cancer status and stage, BMI, bra cup size, smoking status, incision and reconstruction type, and need for chemo or radiation therapy were recorded. BMI was categorized into 3 groups: low (<18.5), normal-overweight (18.5-30), and obese (>30). Chart review was performed to evaluate need and reason for reoperation.   Fisher’s exact test was performed to evaluate BMI and reoperation rates using STATA 14.1 with significance set at a p-value <0.05.

Results

Of 211 pts who underwent 370 NSMs, 6.6% (14/211) had a low BMI, 88.2% (186/211) had normal to overweight BMI, and 5.2% (11/211) were obese (BMI>30). Of 370 NSMs, reoperation was required in 12.4% (46/370): 8.1% (30/370) for nipple/skin necrosis and 4.3% (16/370) for infection. Among pts with low BMI, 21.4% (3/14) required reoperation for either skin or nipple necrosis compared to 16.1% (30/186) in the normal BMI group and 27.3% (3/11) in the high BMI group (p=0.02). In the pts with low BMI, 2 pts suffered bilateral nipple loss. In the pts with high BMI, 1 pt had bilateral nipple loss. 

Conclusion

Pts with a low BMI are at higher risk for clinically significant skin or nipple necrosis that may require reoperation compared to those who have a normal BMI. These findings also confirmed a higher risk of skin or nipple necrosis requiring reoperation in the high BMI group. A larger sample would be needed to confirm these results, but this should be taken into consideration when counseling pts regarding surgical risk.
 

66.02 Decreased Inpatient Mortality after Hepatic Resection in a State Population

D. A. Hashimoto1, Y. J. Bababekov2, S. M. Stapleton2, I. H. Marks2, K. D. Lillemoe1, D. C. Chang2, P. A. Vagefi1  1Massachusetts General Hospital,Department Of Surgery,Boston, MA, USA 2Massachusetts General Hospital,Codman Center For Clinical Effectiveness In Surgery,Boston, MA, USA

Introduction:  There have been considerable improvements in surgical technique and perioperative care in the last decade with respect to hepatic resection for hepatobiliary diseases.  As a result, decreased post-operative mortality has been described at the institutional level. However, inpatient mortality trends following hepatic resection have yet to be assessed on a population level.

Methods:
The New York (NY) Statewide Planning and Research Cooperative System (SPARCS) inpatient database was utilized. All patients over the age of 18 years who underwent wedge hepatectomy or lobectomy from 2000-2014 were included. Trauma and recipient hepatectomy were excluded. Adjusted analysis accounted for age, race, payer status, Charlson Comorbidity Index (CCI), cirrhosis, viral/alcoholic hepatitis, hepatic malignancy (primary vs. secondary tumor), need for biliary-enteric reconstruction, and hospital hepatectomy volume.

Results:

A total of 13,467 hepatectomies were performed from 2000-2014 in the state of NY with a mean inpatient mortality of 2.35% (± 15.1% SD). Of these, 86.6% of hepatectomies were performed at academic centers (hospitals with a surgical residency). Inpatient mortality decreased from a rate of 3.69% in 2000 to 1.98% in 2014 (p<0.0001). Adjusted analysis demonstrated a decreasing trend in mortality from 2000 to 2014 with sustained significance reached in 2009 (OR 0.29, p=0.001) (Figure 1).

Subset analysis revealed similar findings for patients in academic centers, with secondary tumors, or with CCI>3 (all p<0.001). Independent predictors of mortality included age>70 years, male gender, Medicare payer status, primary liver tumor, and need for biliary-enteric reconstruction. Hepatectomy at an academic center (OR 0.62, p=0.002) and female gender (OR 0.67, p=0.001) were protective against mortality.

Conclusion:
This study demonstrates at the state population level that inpatient mortality after hepatectomy has improved over the time period 2000-2014. Increased survival may be due to a combination of advancements in operative and perioperative care. In-depth analyses of surgical care at hospitals in NY may reveal state wide quality improvement practices that led to reduced inpatient mortality after hepatic resections. Such measures could serve as a model for other health systems.

60.09 NF-kB in Stroma Promotes Cancer Growth by Protecting Against Immune Mediated Cytotoxicity

B. Giri1, B. Garg1, S. Modi1, V. Sethi1, S. Ramakrishnan1, S. Banerjee1, A. Saluja1, V. Dudeja1  1University Of Miami,Miami, FL, USA

Introduction: While the role of NFκB in tumor cells in cancer cell growth and metastases is well established, the impact of NFκB in tumor stroma on pancreatic cancer growth is unknown. We sought to understand the effect of depletion of p50 subunit of NFκB in the stromal cells on pancreatic cancer growth and metastases.  

Methods: Pancreatic cancer cells isolated from spontaneous tumors developing in Pdx1-Cre;K-Ras+/LSLG12D;p53R172H/+ (KPC) mice were surgically co-injected with either wild-type (WT) or p50-/- (thus lacking p50 subunit of NFκB) pancreatic stellate cells (PSC). The impact of lack of p50 subunit of NFκB on tumor growth and metastases was measured. This model was repeated in athymic nude mice lacking a functional immune system to assess the effect of p50-/- PSC on tumor cells in the absence of adaptive immunity. The impact of deletion of p50 subunit of NFκB in stroma cells on growth of melanoma (B16-F10) and Lung Cancer (Lewis Lung Cancer) was also evaluated. Immune infiltration in the ex-vivo tumor samples were analyzed by flow cytometry.

Results:Cancer cells co-injected with p50-/- stellate cells formed smaller tumors compared to WT PSC. This was also observed in B16-F10 as well as the Lewis Lung Cancer model. On analysis by flow cytometry, tumors co-injected with p50-/- stellate cells had a higher infiltration of CD8+ cells. Interestingly, the tumor inhibitory effect of p50-/- stellate cells was lost when KPC cells were grown with p50-/- PSC in immuno-deficient athymic nude mice suggesting that stromal cells in the presence of p50 -/- may function to protect cancer cells from an immune mediated attack.

Conclusion:Tumor stroma protects cancer cells from immune-mediated cytotoxicity in NFκB dependent fashion. Developing strategies to downregulate NFκB in tumor stroma may lead to an anti-tumor effect, alone or in combination with immunomodulatory strategies. 

 

60.10 The prognostic value of pseudomyxoma peritonei tumor infiltrating T lymphocytes

C. Boutros1,2, M. Bedra2, J. Emel2, O. Ioffe3, N. Hanna1, N. Espat4, S. Katz4  1University Of Maryland School Of Medicine,Surgery,Baltimore, MD, USA 2University Of Maryland Baltimore Washington Medical Center,Tate Cancer Center,Glen Burnie, MD, USA 3University Of Maryland School Of Medicine,Pathology,Baltimore, MD, USA 4Roger Williams Medical Center,Surgery,Providence, RI, USA

Introduction: Tumor infiltrating lymphocytes (TIL) are independent predictors of survival for numerous metastatic solid tumors. The biologic significance of TIL in pseudomyxoma peritonei (PMP) has yet to be elucidated.

Methods: We accessed our tissue bank for surgical patients after cytoreductive surgery and heated chemotherapy for PMP. Immunohistochemical staining (IHC)for CD3, CD4, CD8 and FOXP3 was correlated with clinicopathologic factors including preoperative CEA and PCI. Markers were correlated with survival using the median cell count as the cutoff point for LOW and HIGH groups.

Results: Of 50 patients, 31 had suitable tissue for IHC analysis. The mean age was 59, preoperative CEA 44.0 and PCI score 17. Median survival was 45 months. CD3, CD4, and CD8 density did not correlate with patient survival. Similarly, preoperative CEA or PCI score alone were not significant predictors of survival. FOXP3, a marker of suppressive regulatory T cells (Treg), was a strong predictor of 5-year survival (82% FOXP3LOW vs 28% FOXP3HI , p=0.001).  FOXP3/CD4 ratio predicted of 30 and 60-month survival (80.5% for LOW vs 31% for HIGH, p=0.038 at 30 months, p=0.004 at 60 months). Similarly, FOXP3/CD8 ratio was found to be a predictor of 60 months survival (77% for LOW, vs 33% for HIGH;p=0.023). Low CD8/CD4 ratio correlated with high preoperative CEA and high PCI score (p=0.01). Patients with the combination of low CEA, low PCI score, and high CD8/CD4 ratio had longer survival times (p=0.01  )

Conclusion: TIL proved to be significant predictors of outcome in this small group of patients. A high number of tumor infiltrating suppressive FOXP3+ Treg predicted shorter survival time, suggesting that immunotherapeutic approaches may benefit PMP-PC patients.

 

60.08 Inhibition of Apelin Receptor Signaling Decreases Cholangiocarcinoma Growth in a Xenograft Model

C. Hall1, L. Ehrlich2, T. Shepperd2, A. O’Brien2, G. Alpini2, S. Glaser2, T. C. Lairmore1  1Scott & White Healthcare,Temple, Texas, USA 2Texas A & M Health Science Center College Of Medicine,Temple, TX, USA

Introduction:
Cholangiocarcinoma (CCA) is a malignancy of the intrahepatic and extrahepatic biliary epithelium that is associated with low five-year survival despite multidisciplinary treatment strategies. Tumor angiogenesis correlates with CCA progression, metastases, and patient survival. The apelin receptor (APLNR), which is activated by the apelin peptide, is a G-protein coupled receptor that has been implicated in the growth and angiogenesis of other malignancies, such as colon, breast, prostate and hepatocellular carcinoma, but has not been studied in CCA. The purpose of this study is to quantify APLNR expression in CCA, characterize the proliferative and angiogenic effects of receptor activation, and determine if inhibition of the APLNR axis can inhibit tumor growth in a murine xenograft model. 

Methods:
In vitro, CCA cell lines (CCLP, HuH-28, HuCCT-1, SG231, TFK-1 and Mz-ChA-1) and benign cholangiocytes (H69) were used to measure the expression of apelin and the APLNR via flow cytometry, ELISA and immunofluorescence. Immunohistochemistry (IHC) and qPCR was used to measure APLNR expression in human CCA tissues. Mz-ChA-1 cells were treated with increasing concentrations of apelin and ML221, an APLNR antagonist. Expression of proliferative (Ki-67 and PCNA) and angiogenic (VEGF-A, ANG1, ANG2) genes were measured via qPCR. Phosphorylation of the ERK1/2 pathway, a known pathway for cholangiocyte proliferation, was measured using flow cytometry and immunoblots. In vivo, Mz-ChA-1 cells were injected into the flanks of NU/NU immunocompromised mice, which were treated with ML221 (150 ?g/kg) via tail vein injection for 4 weeks. 

Results:
APLNR expression and apelin secretion was upregulated in human CCA cells and tissues compared to benign controls. In vitro, treatment of Mz-ChA-1 cells with apelin increased proliferation and angiogenesis via activation of the ERK1/2 pathway in a dose-dependent response, whereas, ML221 inhibited these affects. Treatment of Mz-ChA-1 cells with apelin also increased expression of the apelin gene, suggesting an autocrine/paracrine mechanism of receptor activation. Treatment of CCA tumors in NU/NU mice with ML221 significantly decreased tumor growth in the xenograft model (Figure 1). 

Conclusion:
APLNR is increased in CCA tissues and the autocrine/paracrine effects of APLNR receptor signaling regulate tumor growth and angiogenesis, both in vitro and in vivo. Inhibition of the APLNR axis decreases tumor growth in our xenograft CCA model. Targeting APLNR signaling has the potential to serve as a novel, tumor directed therapy for CCA by inhibiting cell proliferation and angiogenesis.  
 

60.07 Tumor Treating Fields are Effective in Temozolomide Resistant Glioblastoma Cancer Stem Cells

J. K. Strebe5, P. A. Clark5, C. Pasch2, D. A. Demings2,3,4,8, H. I. Robins2,3,6,7,8, J. S. Kuo5  2University Of Wisconsin School Of Medicine And Public Health,Carbone Cancer Center,Madison, WI, USA 3University Of Wisconsin School Of Medicine And Public Health,Division Of Hematology And Oncology, Department Of Medicine,Madison, WI, USA 4University Of Wisconsin School Of Medicine And Public Health,McArdle Laboratory For Cancer Research, Department Of Oncology,Madison, WI, USA 5University Of Wisconsin School Of Medicine And Public Heath,Department Of Neurological Surgery,Madison, WI, USA 6University Of Wisconsin School Of Medicine And Public Health,Department Of Neurology,Madison, WI, USA 7University Of Wisconsin School Of Medicine And Public Health,Department Of Human Oncology,Madison, WI, USA 8William S Middleton Memorial Veterans Hospital,Madison, WI, USA

Introduction:  Tumor Treating Fields (TTFs) are a novel antimitotic, non-invasive, externally applied cancer treatment being developed for use against many human solid cancers. Glioblastoma (GBM) is the most frequently diagnosed adult brain malignancy, with a median survival of less than 2 years after the standard therapeutic regimen of maximal surgery followed by radiation and temozolomide (TMZ) chemotherapy. In a Phase 3 clinical trial (NCT#00916409), addition of TTFs to current therapies significantly increased overall and progression free survival for newly diagnosed GBM. TTFs are FDA-approved for newly diagnosed and recurrent GBM. We report the first study of TTFs alone, or combined with TMZ, against GBM stem-like cells (GSC) expressing different levels of the therapy resistance DNA repair enzyme, O-6-methylguanine DNA methyl-transferase (MGMT), to test the hypothesis that TTF is effective against TMZ-resistant GSC.

Methods:  Effects of TTFs and TMZ were studied in the patient-derived 22 GSC (MGMT-expressing, TMZ resistant) and 33 GSC (non-MGMT-expressing, TMZ sensitive) cell lines with continuous application of in vitro TTFs at varying frequencies to GSC cultures using the Inovitro system (Novocure Ltd). The effects of varying doses or frequencies of TMZ, TTFs, and combined TMZ+TTFs on GSC proliferation and sphere-forming ability were analyzed. 

Results: We have previously determined in vitro that 200 kHz is the optimal TTF frequency to inhibit GSC proliferation, identical to the clinical trial frequency. At 200 kHz, TTFs significantly inhibited proliferation (22 GSC: 61±10.8%; 33 GSC 56±9.5%; p<0.05) and clonogenic tumor sphere formation (22 GSC: 38±2.6%; 33 GSC: 60±7.1%; p<0.05) in both TMZ-resistant and TMZ-sensitive GSC subtypes. In combination, TTFs and TMZ (at IC25, IC50, IC75 concentrations) showed an additive interaction.

Conclusion: This is the first study to directly demonstrate that TTFs can overcome TMZ-resistance in GBM cancer stem cells (GSC), and reports the effects of TTFs on GSC proliferation and clonogenic tumor sphere formation with equivalent effectiveness against both therapy resistant and sensitive GSC subtypes (+/- MGMT expression). The combination of TTFs and TMZ was additive, and supports the hypothesis that TTF acts via a mechanism independent of TMZ-mediated DNA alkylation. Further study of TTFs in additional human cancers and in potential combination with other therapeutics will be needed to optimize TTFs as a new therapy option for human cancer. 

60.06 Brachytherapy with concurrent chemotherapy treatment for anal cancer in transgenic mice

H. Sleiman1, L. Meske1, W. Culberson2, J. Micka2, E. Carchman1  1University Of Wisconsin,General Surgery,Madison, WI, USA 2University Of Wisconsin,Department Of Medical Physics,Madison, WI, USA

Introduction: Chemoradiotherapy is the current standard of care for patients with anal cancer1. The 5 year survival rates of patients with localized disease after chemoradiotherapy is approximately 65%, with 1/3 of these patients developing disease recurrence. The survival rates are significantly less in patients with locally advanced or metastatic disease treated with standard of care. Finally, the toxicity associated with chemoradiotherapy regardless of stage, is not insignificant with 30% of the patients requiring colostomy creation due to therapy related side-effects.  Unfortunately there have been no changes in the treatment for anal cancer over the last 4 decades, largely in part to a lack of preclinical models.  This project describes the first mouse preclinical model for anal cancer therapy with treatment responses mirroring those seen in humans.

Methods: K14E6/E7 (expressing HPV-16 oncoproteins in their epithelium) mice were utilized and anuses treated with 7,12-Dimethylbenz[a]anthracene (DMBA) topically until squamous cell carcinoma of the anus developed. The study contained four treatment groups: no-treatment controls (7 mice), radiotherapy alone-2 Gy (5 mice), chemotherapy alone- mitomycin C and 5-flourauracil (3 mice), and chemoradiotherapy (4 mice). The Xoft® Axxent eBx® system, consisting of an x-ray tube within a cone-shaped skin applicator, was used to irradiate anal tumors to a depth of 3mm at a known absorbed dose.  Anal tumors were measured at three dimensions (mm3) on a daily basis until mice died or met criteria to be removed from the study (22-day treatment time period). All mice tumor sizes were normalized to 0 at day 0 and changes in tumor size over time noted. Groups were then compared using one-way ANOVA.

Results:  Treatment Response: Within 16 days of initiating chemoradiotherapy there was noted to be a significant treatment response compared to no-treatment controls (p-value<0.05). This treatment response was noted, to a lesser degree, in mice that recieved chemotherapy alone.  Please refer to graph 1 for average tumor volume changes over time. Survival Times: Mice that received no treatment survived on average 30 days with signficant growth rates of anal tumors. Mice that received radiation alone survived on average 22 days without significant growth rates of anal tumors, but died of complications due to tumors outside of the anus. Mice that received chemotherapy or chemoradiotherapy survived to the 48 day endpoint.

Conclusion:We have generated a HPV mouse model of anal cancer treatment that mimics responses seen in the humans with chemoradiotherapy. This model can be utilized to test new therapies with the hope of improving response rates and decreasing toxicity in anal cancer treatment.

 

60.05 Minnelide Synergizes with TRAIL Against Pancreatic Cancer

S. Modi1, B. Giri1, V. Sethi1, B. Garg1, J. George1, S. Banerjee1, V. Dudeja1, A. K. Saluja1  1Sylvester Cancer Center,Surgery,Miami, FL, USA

Introduction: Pancreatic cancer is an aggressive malignancy. Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) has been shown to cause cancer cell death in multiple cancers with minimal toxicity to un-transformed cells. Unfortunately, pancreatic cancer (PDAC) is resistant to TRAIL. We have previously shown that triptolide (TPL), a diterpene triepoxide isolated from a Chinese herb and its water soluble pro-drug Minnelide, is effective against pancreatic cancer. Minnelide is currently undergoing Phase-I trials against advanced GI malignancies. The aim of the current study is to evaluate the anti-tumor efficacy of lowered doses of Minnelide in combination with TRAIL in multiple models including an immunocompetent and stroma rich mouse model of pancreatic cancer. 

Methods: The effect of TRAIL (0-40ng/ml), low dose of TPL (50nM), or combination of TRAIL and TPL on the viability (WST-8 assay) and apoptosis (cleaved caspase-3 and cleaved PARP levels using flow cytometry) of human pancreatic cancer cell lines (S2-VP10) and human pancreatic stellate cells (hPSCs) was measured. To evaluate efficacy of combination therapy in animal models of pancreatic cancer, subcutaneous xenograft model in athymic nude mice, subcutaneous syngeneic model in C57BL/6J wild type mice and a novel tumor implantation model in which 3 mm3 pieces of KPC tumors were implanted in pancreas of C57BL/6J wild type mice. The animals were randomized into four groups and treated with Minnelide (0.21 mg/kg/day ip), TRAIL (20mg/kg 3Xweek ip) or combination of the two for 4-6 weeks. At the end of 4-6 weeks, tumors were harvested, and tissues were used for various experiments. The animal doses of Minnelide correspond to the clinically relevant and well tolerated doses from the Phase-I trial.

Results:Combination of TRAIL 1.25ng/ml and TPL 50nM decreased the viability of S2VP10 (viability, % of Control, 25±4.6% after 24h of treatment) and significantly increased cleaved caspase-3 and PARP levels suggesting the activation of apoptotic cell death, whereas TRAIL alone did not influence viability of these cancer cells (viability, % of Control, 96±8.3% at 24h). TRAIL alone caused a downregulation of DR5 receptors while addition of TPL abrogated this effect. In the xenograft SC model, Minnelide and TRAIL alone did not have any effect on tumor size while combination of the two resulted in marked decrease in tumor growth. In the tumor implantation model, combination of low doses of Minnelide and TRAIL markedly inhibited tumor burden when compared to Minnelide or TRAIL alone. Tumor weights (gm, mean±SEM) after 3 week of Rx: Saline-1.63±0.1; Minnelide-1.15±0.02; TRAIL-1.54±0.04; Minnelide+TRAIL-0.38±0.03. TUNEL staining showed significantly increased apoptosis in the combination arm.

Conclusion:Minnelide synergizes with TRAIL therapy and reverses the TRAIL resistance in multiple models of pancreatic cancer including immunocompetent stroma rich model. 

 

60.04 BIRC5 Is A Biomarker for Early Detection of PDAC in CRISPR-Cas9 Engineered Pancreatic Organoids

S. Liu1, J. Yu1, R. Sanchez1, E. Rozengurt2, F. Brunicardi1  1David Geffen School Of Medicine, University Of California At Los Angeles,Department Of Surgery,Los Angeles, CA, USA 2David Geffen School Of Medicine, University Of California At Los Angeles,Division Of Digestive Disease,Los Angeles, CA, USA

Introduction: Recent advances in CRISPR-Cas9 gene editing and organoid culturing are leading to pancreatic cancer (PDAC) tumor models with unprecedented speed and precision by introducing driver mutations such as KrasG12D, P53 mutation, which will hopefully identify early detection PDAC biomarkers. Over-expression of BIRC5 has been found in early stage of PDAC in human specimens and is a promising candidate for an early detection biomarker. The purpose of this study is to study BIRC5 as an early detection biomarker by taking the advantage of CRISPR-Cas9 and organoid techniques to develop an early onset pancreatic 3D tumor model.

Methods:  Three-dimensional culture conditions were optimized to maintain mouse pancreatic ductal progenitor organoids (ORGs). sgRNA was well designed and cloned into a GFP-tagged lentiviral vector to generate two vectors: Lenti-sgKrasG12D-GFP and Lenti-sgKrasG12D-sgp53KO-GFP. Co-transfection was performed using Lenti-sgRNA-GFP and Lenti-Cas9-2A-tdTomato (tdT) to infect ORG. KrasG12D and p53 mutations were confirmed by genome DNA sequencing. BIRC5 expression in ORGs and human specimens was measured using western blot and immunofluorescence assay. Knockdown of Kras and overexpression of wild type p53 was carried out. A reporter assay using BIRC5-GLuc was performed in these ORGs. 

Results:Immunofluorescence demonstrated that BIRC5 was expressed in PanIN3 and PDAC specimens.   ORGs were established in matrigel with additional growing factors in culture media. GFP (green), tdT (red) or GFP/tdT double staining was observed in ORGs following co-infection. Tumoroid ORGs were observed in CRISPR-Cas9 engineered ORGs displaying GFP/tdT double staining in 3-D culturing with either GFP or tdT only (Fig). The expression of GFP and tdT were further confirmed in the cryosection and paraffin sections. KrasG12D mutation only resulted in very limited expression of BIRC5 in ORGs, however KrasG12D/p53KO mutations markedly increased BIRC5 expression. Knockdown of KrasG12D expression did not alter BIRC5 levels, however, restoration of wild type P53 expression resulted in significant reduction of BIRC5 levels in ORGs (p<0.05). Furthermore, BIRC5-GLuc reporter assay revealed GLuc expression only in the ORGs with both KRAS and P53 mutations. 

Conclusion:CRISPR-Cas9 engineering of pancreatic ductal organoids with KRAS/P53 driver mutations resulted in a tumoroid ORGs associated with BIRC5 overexpression. BIRC5 was over-expressed in PanIN3 and PDAC. These data support the hypothesis that BIRC5 is biomarker for early detection of PDAC.

 

60.03 Enrichment of Polymerase Theta (POLQ) Single Nucleotide Polymorphisms in Papillary Thyroid Carcinoma

T. D. Murtha1, R. Korah1, T. Carling1  1Yale University School Of Medicine,New Haven, CT, USA

Introduction:  Recent comprehensive genetic analyses have demonstrated a high prevalence of gene fusions–suggestive of defective DNA break repair–in papillary thyroid carcinoma (PTC). Polymerase theta (POLQ) is an A-family mammalian polymerase that is unique in mediating replication-independent DNA repairs, including single and double strand DNA breaks. Differential expression of POLQ has been shown to correlate with poor outcomes in breast, ovarian, and lung cancer. Because of the high concentration of hydrogen peroxide in thyroid follicles and the consequent oxidation-induced single and double strand breaks in DNA, we hypothesized that genetic polymorphism of the DNA repair enzyme POLQ may play a role in predisposing follicular cells to de novo mutations and potentially tumorigenesis. To identify novel mutations and enrichment of SNPs, we performed classical Sanger sequencing of the exonic regions of POLQ in a large cohort of sporadic PTCs. 

Methods:  Genomic DNA was isolated from 51 histologically confirmed PTC specimens and matched adjacent non-tumor thyroid tissue. Genetic sequencing via capillary electrophoresis (Sanger method) was performed to detect novel variants and SNPs for all 30 exons and exon/intron splice sites of the POLQ gene. Discovered nonsynonymous and missense SNPs, and the corresponding amino acid substitutions, were analyzed using multiple bioinformatic algorithms to determine if alterations were deleterious to protein integrity or function. Novel variants were annotated and SNP frequency was compared to The Exome Aggregation Consortium (ExAC) database.

Results: Fourteen germline variants were confirmed in tumor and corresponding matched adjacent non-tumor thyroid tissue. Eight of those germline variants (57%) were determined to be disease-causing by mutation prediction algorithms. In the C-terminal polymerase domain (amino acids 2060–2590), there was an increased frequency of germline SNPs compared to the comprehensive population SNP database ExAC. The recurrent SNP rs532411, previously found to be significantly associated with breast cancer, was found in 22% (11/51) of PTC specimens while its frequency in the population is 6.5% (P < .0001, chi-squared test). We also identified 4 novel variants not previously reported.

Conclusion: The unique contribution of POLQ in DNA repair via non-homologous end-joining of double stand breaks makes it a critical regulator of genomic integrity in the hydrogen peroxide-rich thyroid gland. Sequencing demonstrates enrichment of multiple SNPs in the polymerase region of POLQ, potentially increasing the probability of carcinogenic point mutations and gene fusions. While expanded genome-wide association studies (GWAS) are essential for confirming the relationship, the current study offers preliminary information suggesting a role for POLQ SNPs in genetically predisposing individuals to PTC.

60.02 Utility of Circulating Tumor Cells for Preoperative Prediction of Micrometastatic Pancreatic Cancer

C. M. Court1,4, J. S. Ankeny1,4, S. Sho1, S. Hou1, P. Winograd1, Q. Li1, M. Song1, T. R. Donahue1,4, O. J. Hines1, H. A. Reber1, Z. A. Wainberg3, H. R. Tseng4, J. S. Tomlinson1,4  2University Of California – Los Angeles,Molecular And Medical Pharmacology,Los Angeles, CA, USA 3University Of California – Los Angeles,Hematology/Oncology,Los Angeles, CA, USA 4VA Greater Los Angeles,Surgery,Los Angeles, CA, USA 1University Of California – Los Angeles,Surgery,Los Angeles, CA, USA

Introduction: Current preoperative evaluation and staging of patients with pancreatic ductal adenocarcinoma (PDAC) is hampered by the limited sensitivity of cross-sectional imaging for micrometastatic disease. Furthermore, the majority of patients who undergo resection ultimately succumb to metastatic disease, suggesting that current staging systems are likely routinely understaging patients. Our goal was to investigate the utility of circulating tumor cells (CTCs) as a preoperative predictor of metastatic disease and postoperative outcomes.

Methods:  A total of 49 patients were taken to the operating room for attempted pancreatic resection and enrolled in the study. 15 (30.6%) had undergone neoadjuvant therapy but were deemed to be surgical candidates based on post-treatment imaging. Four milliliters of venous blood (VB) was evaluated for the presence and number of CTCs using the microfluidic NanoVelcro chip. CTCs were defined by immunocytochemical staining (CK+ or CEA +, CD45-, DAPI+). CTC number was then correlated with both surgical outcomes and standard clinicopathologic findings.

Results: Of the 49 PDAC patients taken to the operating room, 11 (22.4%) were found to have metastatic disease intraoperatively, 6 with liver metastases and 5 with peritoneal metastases. CTC presence and number per 4 mL VB were found to correlate with stage and distinguished patients with resectable disease from those with metastatic disease. CTCs were present in all (100%) of the 11 patients with metastatic disease versus only 63% of those with resectable PDAC and at significantly higher levels (Average CTCs/4mL VB- 11.5 vs. 1.7, p<0.01). Using a cutoff of ≥3 CTCs/4mL VB, CTCs were able to distinguish patients with metastatic disease from those with resectable cancer with a sensitivity of 90.9%, specificity of 84.2%, PPV of 62.5%, NPV of 97.0%, and AUROC of 0.898 (95% CI = 0.796 – 0.999, p < 0.001). Of note, CA19-9 levels did not statistically differ between the groups (p=0.88). Furthermore, higher CTC counts correlated with worse recurrence-free and overall survival for both the entire cohort as well as the group of patients who successfully underwent an operation.

Conclusion: In this small prospective study, preoperative CTC enumeration demonstrated a correlation with the presence micrometastatic disease not detected by preoperative cross sectional imaging. Larger studies with longer follow-up are needed to firmly establish CTCs as a predictive biomarker in PDAC. 

 

60.01 U1 adaptors suppress the KRAS-cMYC oncogenic axis in human pancreatic cancer xenografts

A. T. Tsang1,6, L. Yi1, C. Dudgeon1, X. Yu1, R. Goraczniak5, S. Gunderson3,5, D. R. Carpizo1,2,4  6Mount Sinai St. Luke’s Roosevelt General Surgery Residency Program,New York, NY, USA 1The Cancer Institute Of New Jersey,New Brunswick, NJ, USA 2Rutgers University Robert Wood Johnson Medical School,Surgery,New Brunswick, NEW JERSEY, USA 3Rutgers University,Department Of Molecular Biology And Biochemistry,PIscataway, NEW JERSEY, USA 4Rutgers University,Department Of Pharmacology,Piscataway, NEW JERSEY, USA 5Silagene, Inc.,Hillsborough, NEW JERSEY, USA

Introduction:
One of the most significant unmet needs in pancreatic cancer therapy is targeting the most commonly mutated gene, KRAS, and its downstream mediator MYC. Small interfering RNA targeting KRAS has produced potent anti-tumor activity in preclinical studies, but technical difficulties of in-vivo delivery have impeded clinical translation. U1 Adaptors are a novel technology for oligonucleotide-mediated gene silencing that acts by blocking polyadenylation of messenger RNA. They can accommodate extensive covalent modifications for nuclease resistance, targeted delivery and in-vivo imaging without loss of silencing activity, offering important advantages over siRNA and antisense oligos as therapeutic agents. The KRAS-cMYC oncogenic axis plays a key role in the generation of self-renewing metastatic cells, making cMYC an attractive target in KRAS-driven pancreatic cancers. Genetic studies in pancreatic cancer mouse models validate the therapeutic efficacy of silencing KRAS and MYC expression.

Methods:
Candidate U1 Adaptors targeting KRAS were screened in vitro using the human pancreatic cancer cell line MIAPaCa2 (KRAS G12D mutant). The best Adaptors were applied in cell growth inhibition assays over 10 days in multiple pancreatic cancer cell lines. They were then tested for efficacy in mice bearing subcutaneous MIAPaCa2 xenograft tumors. For in-vivo delivery, Adaptors were covalently linked to a cyclic RGD-motif peptide (cRGD), a targeting ligand for integrin receptors expressed on tumor cells and endothelia, or alternately, internalizing RGD (iRGD), a variant peptide that triggers endothelial permeabilization and internalization by cells through neuropilin-1 binding. The cRGD- and iRGD-conjugated KRAS Adaptors were administered by tail vein injections twice weekly for 3 to 4 weeks once tumors reached 20mm3.. In parallel, U1 Adaptors targeting cMYC were screened in B-cell lymphoma lines and tested for efficacy in mice bearing MIA-PaCa-2 xenograft tumors using the same method.

Results:
The best KRAS Adaptors reduced KRAS mRNA expression by up to 76% – as effectively as an siRNA control. Knockdown of KRAS protein expression and its downstream effectors was confirmed by western blot. Cell growth inhibition was demonstrated for MIAPaCa2 and other established human pancreatic cancer cell lines in vitro. The potency of inhibition was dependent on mutant KRAS. Over a series of in vivo mice experiments, MIAPaCa2 xenograft tumor growth was inhibited by averages of 68% to 93% by cRGD- and iRGD-conjugated KRAS Adaptors as compared to vehicle-only controls. Tumor stasis or regression occurred in some treated mice. Remarkably, cMYC Adaptors were similarly effective in suppressing xenograft tumor growth.

Conclusion:

U1 Adaptors can successfully target human KRAS and cMYC in vivo. These results support the continued development of U1 Adaptor technology as a strategy for therapeutic suppression of KRAS, cMYC and possibly other oncogenes in pancreatic cancer.

 

59.06 Strategies to Induce Anti-Tumor Immunity in Colorectal Liver Metastases

N. M. Kunda1, J. Qin1, G. Qiao1, B. S. Prabhakar2, A. V. Maker1,2  1University Of Illinois At Chicago,Division Of Surgical Oncology, Department Of Surgery, College Of Medicine,Chicago, IL, USA 2University Of Illinois At Chicago,Department Of Microbiology & Immunology, College Of Medicine,Chicago, IL, USA

Introduction:
Anti-tumor immune responses have been shown to improve outcomes in patients with advanced stage colon cancer.  Immunogenic cell death (ICD) is a specific mechanism of drug-induced apoptosis that can stimulate an anti-tumor immune response through activation of specific T-cell responses.  Mitoxantrone (MTX) is an anthracenedione antineoplastic agent known to trigger ICD, however, it has not been evaluated as a potential immunotherapeutic potentiator in colon cancer or its metastases.  

Methods:
Murine colon cancer CT-26 cells were treated with 1 μM MTX. For cell cycle studies, cells were fixed with 70% ethanol and DNA staining was performed using DAPI. DNA content was measured with FACS and cell cycle distribution was analyzed.  Treated colon cancer cells were then injected subcutaneously into the right flank of Balb/c mice on days 0 and 7. On day 14, isolated colorectal liver metastases were induced using our well-established model of intrasplenic tumor cell injection of untreated CT-26 cells. All animals were sacrificed on day 28 after establishment of liver metastases.

Results:
At clinical concentrations of MTX, the fraction of cells in the G2-M phase increased from 15.6%-38.8%, and the fraction of cells in the G1 phase decreased correspondingly from 62.6% to 32.8%, consistent with cell cycle growth arrest. The fraction of cells in the sub-G1 phase increased from 0.8% to 11.2%, consistent with DNA degradation and apoptosis.  Liver metastatic tumor burden and average liver weights were significantly decreased in the MTX-treated group compared to control vaccinated animals (4.6g vs 1.8g, p<0.05). The percentages of infiltrating NK cells and activated T-cells in metastatic liver tumors were significantly higher in the MTX-treated group compared to controls (p<0.05). Splenocytes from treated animals also trended towards increased populations of CD3 cells (7.9% vs 17.9%), NK cells (3.6% vs 6.3%), & CD8 cells (24.1% vs 28.6%) compared to control-treated animals. 

Conclusion:
Treatment of colon cancer cells with MTX induced cell cycle arrest, and vaccination of animals with MTX-treated colon cancer cells induced significantly increased immune cell infiltration and decreased tumor burden in distant colorectal liver metastases.  Evaluation of MTX as a stimulator of immunogenic cell death and as a strategy to induce anti-tumor immunity for the treatment of advanced stage colorectal cancer is warranted. 
 

59.05 Interleukin-22 Promotes Chemoresistance in Pancreas Cancer by Enhancing DNA Damage Repair

J. Lazarus1, M. Perusina Lanfranca1, W. Wang1, T. Maj1, W. Zou1, T. Frankel1  1University Of Michigan,Ann Arbor, MI, USA

Introduction:
Pancreatic cancer (PDAC) is the 3rd leading cause of cancer death in the United States.  Patients diagnosed with PDAC have a poor 5-year survival which has not significantly improved in decades. Chemotherapy, including platinum based agents, is often ineffective and the development of chemo-resistance is common.  Recent data has shown that interleukin-22 (IL-22), a cytokine produced by immune cells which acts primarily on epithelial cells, is associated with a worse prognosis in PDAC. We hypothesize that IL-22 enhances chemoresistance leading to the observed impairment in survival.

Methods:
PDAC cells were incubated with and without IL-22 for 3 days followed by cisplatin treatment for 24 hours.  Cell death was determined by propidium iodide (PI) staining.  Apoptosis was assessed by annexin V staining by flow cytometry and immunoblotting for cleaved caspase 3 (cCASP3). Cisplatin mediated DNA damage was evaluated by western blot for the DNA damage marker γH2AX. Genes and proteins involved in apoptosis, DNA damage recognition and repair were assessed by rtPCR and western blotting, respectively.

Results:
Cells pretreated with IL-22 exhibited less platinum chemotherapy-mediated cell death compared to controls as determined by PI staining. In particular, IL-22 decreased the number of cisplatin-induced apoptotic cells and inhibited cCASP3. To investigate the mechanism of IL-22 protection, we first examined differences in apoptotic related gene expression and determined no appreciable differences in Bcl-2, Bcl-xL, caspase 3, and BAX. However, we found cisplatin-induced γH2AX was decreased significantly following IL-22 pre-treatment, suggesting IL-22 controls cisplatin resistance at the DNA damage response level. The role of IL-22 in DNA damage response was investigated by examining expression of genes involved in homologous recombination, non-homologous end-joining, base excision repair and nucleotide excision repair. Data revealed that IL-22 increased gene expression of BRCA2 and PARP in cells also treated with cisplatin.

Conclusion:
Elevated IL-22 is associated with a poor prognosis in pancreas cancer. We determined that IL-22 protected PDAC cells from platinum induced cell death by enhancing DNA damage repair by homologous recombination and base excision repair as evidenced by decreased γH2AX and up-regulation of BRCA2 and PARP.  Strategies to derail DNA damage repair in tumors with a high level of IL-22 may improve sensitivity to chemotherapy and subsequently survival.
 

59.04 Importance of Lymphodepletion prior to Cancer Immunotherapy with Specific T Cell Subsets

M. V. Beems1,2, A. Contreras1, T. K. Luther2, A. Tatar1, P. Srinand1, S. Sen1, C. S. Cho2  1University Of Wisconsin,Department Of Surgery,Madison, WI, USA 2University Of Michigan,Department Of Surgery,Ann Arbor, MI, USA

Introduction:
Adoptive cell transfer (ACT) immunotherapy traditionally involves the infusion of tumor-specific T cells into cancer patients after a lymphodepletion regimen designed to enhance T cell engraftment. Lymphodepletion is a potential source of patient morbidity. Our laboratory and others have shown that ACT using memory T cells is superior to traditional effector T cell-based ACT. Given the innate ability of memory T cells to persist well after antigen encounter, we hypothesized that ACT with memory cells would be less dependent on pre-transfer lymphodepletion.

Methods:
C57BL/6 mice were inoculated with B16GP33 melanoma flank tumors. Half of the mice underwent pre-transfer lymphodepletion using 5Gy total body irradiation. Equal numbers of tumor-specific effector and memory T cells were adoptively transferred. Serial tumor measurements and flow cytometric analyses of tumors, tumor draining lymph nodes (TDLNs), and circulating blood were performed.

Results:
Lymphodepletion significantly reduced tumor growth in all groups. Tumor control was strongest in mice receiving memory T cell ACT with lymphodepletion, with minimal tumor growth seen in this group. Flow cytometric analyses revealed a significantly higher proportion of adoptively transferred cells in the tumors, TDLNs, and circulating blood in mice treated with pre-transfer lymphodepletion. Lymphodepletion benefited memory ACT more than effector ACT in terms of increasing adoptively transferred T cell numbers in all lymphoid compartments.

Conclusion:
Contrary to our hypothesis, lymphodepletion with total body irradiation improved the efficacy of memory ACT to a greater extent than effector ACT. ACT of tumor-specific memory T cells following pre-transfer lymphodepletion may represent a novel modality of immunotherapy for melanoma cancer patients.

57.12 Promoting Clinician Engagement in Tobacco Cessation: A Pilot Study

J. Pollichemi2,3, M. Masika2,3, O. Lucas2,3, B. Bigham1,2, K. Attwood2,3, M. Reid2,3, M. Mahoney2,3, C. Nwogu2,3  1Howard University College Of Medicine,Washington, DC, USA 2Roswell Park Cancer Institute,Buffalo, NY, USA 3State University Of New York At Buffalo,Buffalo, NY, USA

Introduction:
Smoking cessation, regardless of a patient’s prognosis, is important for improving health, quality of life, and reducing comorbidities. Therefore, it is important that clinicians provide smoking cessation services. This study assesses the effectiveness of a 1-hour educational interventional program on clinicians providing smoking cessation services in the thoracic clinic at a comprehensive cancer center.

Methods:
New patients that were current smokers with cancer were identified by a retrospective chart review. In order to establish current clinician behavior, no action was taken to alter practices six weeks prior to the educational intervention. Patients seen in the thoracic clinic during this time were assigned to the ‘before cohort’. Patients seen six weeks after the intervention were assigned to the ‘after cohort’. The cohorts were compared by assessing the number of clinicians that provided the following smoking cessation services: advising patients to quit, counseling patients, and offering pharmacotherapy.

Results:

A total of 257 charts were reviewed: 141 belonged to the ‘before cohort’ and 116 belonged to the ‘after cohort’. Of the ‘before cohort’, 27 were current smokers, 69 were former smokers, 28 never smoked and 17 were undocumented. Of the ‘after cohort’, 23 were current smokers, 61 were former smokers, 27 never smoked and 5 were undocumented. The demographics, comorbidities, and smoking habits did not differ significantly between the two cohorts. There was a trend towards an increase in clinicians providing smoking cessation services after the educational session but it did not reach statistical significance (Figure 1).

Conclusion:

These results indicate that barriers exist for providing smoking cessation services. Perhaps a larger sample size is needed to identify a smaller effect that this educational program had on clinician practices. It is also possible that ongoing education programs are necessary to have an appreciable effect on clinician behavior.

 

[Figure 1:  Smoking cessation services in the pre-intervention versus post-intervention cohorts]

54.03 Utilization of Emergency Department Care by Cancer Patients in the United States

A. A. Shah1,2, S. Zafar1, R. Gray2, B. Pockaj2, E. Cornwell1, L. Wilson1, N. Wasif2  1Howard University College Of Medicine,Surgery,Washington, DC, USA 2Mayo Clinic In Arizona,Surgery,Phoenix, AZ, USA

Introduction:  Utilization of emergency department (ED) services by cancer patients has not been well studied. Our objective with this study was to identify common reasons for ED visits in patients with cancer and identify predictors for subsequent admission.

Methods:  The Nationwide Emergency Department Sample (2009-2012) was queried for patients with a diagnosis of malignant cancers (ICD-9-CM diagnosis codes; 140-208.9, 238.4, 289.8) as a secondary diagnosis. Of these the five most common cancers in the United States, as identified by the American Cancer Society, were identified. Primary diagnosis codes were examined for common reasons for presentation to the ED. Descriptive analysis was then performed to describe patient demographics, payor status, discharge disposition, hospital characteristics and outcomes. Multivariable logistic regression analyses for inpatient admission were used to identify risk factors from among the following domains age, gender, insurance status, income, and year of admission for all cancer patients and for each of the commonest cancers (Table).

Results: A total of 2,279,822 records were analyzed representing 2% of ED visits and weighted to represent 10,178,361 visits nationally. Mean age was 63.9(±17.9) with a slight female dominance. Medicare was the primary payor for 55.6% and Medicaid for 12.5%, whereas 24.1% had private insurance. Of the 5 most common cancers, patient with lung cancer comprised 11.8% of ED visits followed by prostate(6.5%), breast(5.7%), colorectal(4.6%), and bladder(1.9%) cancers. Around 65.0% were admitted to the hospital and 31.1% were discharged from the ED. Geriatric patients and those in the highest income quartile are at higher risk of hospital admission. However, female patients, the uninsured and those visiting on the weekends were less likely to be admitted to the hospital (table). The five most common reasons for ED visits included pneumonia(3.5%), abdominal pain (3.5%), urinary tract infection(2.1%), acute exacerbation of bronchitis(1.7%) and acute kidney injury(1.6%). Mortality was 0.4% and 4.0% in the ED and inpatient, respectively. Amongst the five most common cancers, patients with lung cancers (OR[95%CI]:2.07[2.04-2.10]) had the highest odds likelihood of admission followed by patients with bladder cancer (OR[95%CI]:1.71[1.67-1.75]) and colorectal cancer (OR[95%CI]:1.50[1.47-1.52]).

Conclusion: Cancer patients represent an important patient demographic treated in the ED every year. The results of this study help identify the spectrum of clinical conditions cancer patients present with. Recognizing patients at risk for admission can help expedite ED triage to reduce wait times, ensure timely healthcare delivery and identify potentially avoidable admissions.
 

54.01 Nomogram to Predict Risk for Regional Lymph Node Metastasis for Appendiceal Neuroendocrine Tumors

C. Mosquera1, N. Bellamy1, T. L. Fitzgerald1  1East Carolina University Brody School Of Medicine,Division Of Surgical Oncology,Greenville, NC, USA

Introduction:  Currently, size is the primary factor utilized to determine risk of regional nodal metastasis for Appendiceal Neuroendocrine Tumors (A-NET). Here we validate a nomogram combining depth of invasion and size to predict risk of nodal disease. 

Methods:  Patients with resected A-NET from 2004-2013 were identified in the NCDB. 

Results: A total of 3,269 patients were included. The majority were female (56.9%), white (88.1%), had no nodal metastasis (74.9%), and received colectomy (61.5%). On univariate analysis, risk of nodal metastasis was associated with greater depth of invasion (LP 13.3%, MP 22.5%, TS 60.0%, p<0.0001), tumor size (<1 cm 3.6%, 1-2 cm 19.8%, 2-4 cm 45.6%, > 4 cm 44.1%, p<0.0001), and extent of surgical resection (appendectomy 12.8%, colectomy 30.0%, p<0.0001). On multivariate analysis depth of invasion (LP vs MP OR 1.03 p=0.8924; LP vs TS OR 4.02, p <0.0001), size (<1 cm vs 1-2 cm OR 5.81, p<0.0001; <1cm vs 2-4 cm OR 16.78, p<0.0001; <1 cm vs >4 cm OR 13.02, p <0.0001), and extent of surgical resection (colectomy vs appendectomy OR 2.09, p<0.0001) continued to be significant. On univariate survival analysis of 5-year DSS, depth of invasion (LP 88.5%, MP 84.8%, TS 58.2%, p<0.0001), size (<1 cm 84.5%, 1-2 cm 86.3%, 2-4 cm 81.5%, >4 cm 75.4%, p=0.0004), and extent of surgical resection (appendectomy 85.3%, colectomy 80.7%, p=0.0006) were predictive of survival. On multivariate survival analysis, increased depth of invasion (LP vs MP HR 1.73 p=0.1709; LP vs TS HR 5.63, p=0.0023) and size (<1 cm vs 1-2 cm HR 0.12, p<0.0001; <1cm vs 2-4 cm HR 0.34, p=0.0129; <1 cm vs >4 cm HR 0.21, p=0.0034) were associated with survival, however, extent of surgical resection was not (colectomy vs appendectomy HR 1.86, p=0.1188). A nomogram was created to assess the risk of nodal metastasis determined by size and depth of invasion (see figure). The model accurately predicts risk of lymph node metastasis for A-NET with an area under the Received Operating Characteristic (ROC) curve of 0.77200. In order to eliminate bias of low lymph node retrieval with only appendectomy, a model including only colectomy patients was constructed. All results were similar with the ROC of 0.75301.

Conclusion: This study validates the utility of a nomogram including depth of invasion and size to predict risk of nodal metastasis of A-NET. Given that depth predicts both risk of lymph node metastasis and mortality, consideration should be given to including this data in AJCC T classification. 

51.19 Socioeconomic Disparities Affect Survival in Malignant Ovarian Germ Cell Tumors in AYA Population

L. V. Bownes1, I. I. Maizlin1, K. Gow2, M. Langer5, M. Goldfarb3, M. Raval7, J. Doski6, A. Goldin2, J. Nuchtern4, S. Vasudevan4, E. A. Beierle1  1University Of Alabama at Birmingham,Division Of Pediatric Surgery,Birmingham, Alabama, USA 2University Of Washington,Seattle, WA, USA 3Providence Saint John’s Health Center,Santa Monica, CA, USA 4Baylor College Of Medicine,Houston, TX, USA 5Maine Medical Center,Portland, ME, USA 6University Of Texas Health Science Center At San Antonio,San Antonio, TX, USA 7Emory University School Of Medicine,Atlanta, GA, USA

Introduction: Malignant ovarian germ cell tumors (MOGCT) comprise approximately 5% of primary ovarian malignancies. Although current treatments provide excellent outcomes, survival has been shown to be related to race and age. Socioeconomic (SE) factors have been proposed to affect survival in other cancers, but their effect on survival in MOGCT has yet to be evaluated. Therefore, we examined whether SE status impacted the survival of adolescent and young adult women (AYA) with MOGCT.

Methods: The National Cancer Data Base was used to identify all AYA female patients (14-39 years old) with diagnosis of MOGCT from 1998 to 2012. Three SE surrogate variables were identified: insurance type (private, government, uninsured), median income and percent of people without a high school degree in patient’s ZIP code. Pooled-variance t-tests and χ2 were used to compare tumor characteristics, time from diagnosis to staging and to treatment, and clinical outcome variables within each of the SE surrogate variables, while controlling for the effect of age and race in a multivariate model.

Results: 3125 AYA patients were diagnosed with MOGCT. Controlling for age and race, there were significant differences in tumor stage and size at diagnosis when compared between insurance groups, income, and education quartiles in patients within lower quartiles of all measures having larger and more aggressive tumors (Table). Following diagnosis, there was no significant difference in time to tumor staging between insurance groups (p=0.062), income quartiles (p=0.196) or education level (p=0.417). Similarly, there was no association of insurance (p=0.85), income (p=0.28), or education (p=0.61) levels in time to treatment. No significant difference was found between the groups in type of surgery. Survival analysis demonstrated higher mortality to be associated with lower level of education (p=0.001; Hazard Ratios [HR] = 0.87, 0.52 and 0.39, compared to lowest quartile), income quartile (p=0.002; HR=0.54, 0.39 and 0.34, compared to lowest quartile) and insurance status (p<0.001; HR=0.61 for government insurance, HR=0.46 for private insurance, compared to uninsured). Controlling for stage and size of tumor, the difference in survival loses significance, indicating that the original difference in survival is likely due to disparity in extent of disease at presentation.

Conclusions: Female AYA patients from lower SE status with MOGCT presented with more extensive disease, which translated into lower survival, despite similar treatment patterns and equal time to definitive treatment. The underlying factors resulting in these differences must be further examined as potential targets for improved education and access to initial care.