Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Publication year range
1.
J Am Coll Surg ; 239(1): 9-17, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38445645

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and lethal malignancy. Surgical resection is the only curative modality combined with neoadjuvant chemotherapy to improve survival. Given the limitations of traditional responses such as cross-sectional imaging (CT/MRI) or tumor markers, carbohydrate antigen 19-9 (CA19-9), the 2023 National Comprehensive Cancer Network guidelines included 18 F-fluorodeoxyglucose (FDG)-PET as an adjunct to assess response to neoadjuvant chemotherapy. There are common misconceptions on the metabolic activity (tumor avidity) in PDAC so we aimed to describe the baseline characteristics and use of FDG-PET in a cohort of treatment-naive patients with PDAC. STUDY DESIGN: A single-center retrospective study was conducted capturing all biopsy-proven, treatment-naive patients with PDAC who underwent either baseline FDG-PET/CT or FDG-PET/MRI imaging between 2008 and 2023. Baseline FDG-PET characteristics were collected, including primary tumors' maximum standardized uptake value defined as metabolic activity (FDG uptake) of tumor compared with surrounding pancreatic parenchymal background, and the identification of extrapancreatic metastatic disease. RESULTS: We identified 1,095 treatment-naive patients with PDAC who underwent baseline FDG-PET imaging at diagnosis. CA19-9 was elevated in 76% of patients. Overall, 96.3% (1,054) of patients had FDG-avid tumors with a median maximum standardized uptake value of 6.4. FDG-PET also identified suspicious extrapancreatic metastatic lesions in 50% of patients, with a higher proportion (p < 0.001) in PET/MRI (59.9%) vs PET/CT (44.3%). After controlling for CA19-9 elevation, PET/MRI was superior in detection of extrapancreatic lesions compared with PET/CT. CONCLUSIONS: FDG-PET has significant use in PDAC as a baseline imaging modality earlier neoadjuvant therapy given the majority of tumors are FDG-avid. FDG-PET can identify additional extrapancreatic suspicious lesions allowing for optimal initial staging, with PET/MRI having increased sensitivity over PET/CT.


Subject(s)
Carcinoma, Pancreatic Ductal , Fluorodeoxyglucose F18 , Pancreatic Neoplasms , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals , Humans , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Male , Retrospective Studies , Female , Middle Aged , Aged , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Positron Emission Tomography Computed Tomography/methods , Magnetic Resonance Imaging , Adult , Positron-Emission Tomography/methods , Aged, 80 and over
2.
Ann Surg Oncol ; 30(7): 4377-4387, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36964844

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) requires complex multidisciplinary care. European evidence suggests potential benefit from regionalization, however, data characterizing the ideal setting in the United States are sparse. Our study compares the significance of four hospital designations on guideline-concordant care (GCC) and overall survival (OS). PATIENTS AND METHODS: The Texas Cancer Registry was queried for 17,071 patients with PDAC treated between 2004 and 2015. Clinical data were correlated with hospital designations: NCI designated (NCI), high volume (HV), safety net (SNH), and American College of Surgeons Commission on Cancer accredited (ACS). Univariable (UVA) and multivariable (MVA) logistic regression were used to assess associations with GCC [on the basis of National Comprehensive Cancer Network (NCCN) recommendations]. Cox regression analysis assessed survival. RESULTS: Only 43% of patients received GCC. NCI had the largest associated risk reduction (HR 0.61, CI 0.58-0.65), followed by HV (HR 0.87, CI 0.83-0.90) and ACS (HR 0.91, CI 0.87-0.95). GCC was associated with a survival benefit in the full (HR 0.75, CI 0.69-0.81) and resected cohort (HR 0.74, CI 0.68-0.80). NCI (OR 1.52, CI 1.37-1.70), HV (OR 1.14, CI 1.05-1.23), and SNH (OR 0.78, CI 0.68-0.91) all correlated with receipt of GCC. For resected patients, ACS (OR 0.63, CI 0.50-0.79) and SNH (OR 0.50, CI 0.33-0.75) correlate with GCC. CONCLUSIONS: A total of 43% of patients received GCC. Treatment at NCI and HV correlated with improved GCC and survival. Including GCC as a metric in accreditation standards could impact survival for patients with PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , United States/epidemiology , Pancreatic Neoplasms/surgery , Carcinoma, Pancreatic Ductal/therapy , Texas/epidemiology , Hospitals , Pancreatic Neoplasms
3.
Perm J ; 27(2): 9-12, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36336674

ABSTRACT

Introduction Adenosquamous carcinoma (ASC) of the pancreas is a rare form of pancreatic cancer with a worse prognosis than pancreatic ductal adenocarcinoma. The authors report on a retrospective study of 13 patients diagnosed with ASC in an integrated health care system. Methods A retrospective review was performed of all patients with pancreatic cancer identified between February 2010 and December 2018. Twenty-three patients were diagnosed with pancreatic ASC. Patient demographics, tumor characteristics, treatment modalities, and median survival were evaluated. Results Median overall survival was 8 months (standard devision [SD] = 18.6). Eight out of 13 patients who received surgery upfront had a positive surgical margin (62%). Eleven patients received adjuvant therapy. Median survival for patients who received multimodal treatment was 57 months (SD = 5.7) compared with 2.5 months for patients who received only surgery. Median survival for patients with negative pathologic margins was 17 months (SD = 23.6). One patient was receiving neoadjuvant chemotherapy (6 months into treatment without any evidence of metastatic disease). Discussion The high proportion of positive surgical margins and large tumor size upon presentation suggest that primary tumor downstaging should be considered. The positive results from recent prospective trials on neoadjuvant chemoradiation for pancreatic ductal adenocarcinoma could be a promising foundation of information for the treatment of ASC. Conclusion ASC of the pancreas is an extremely aggressive malignancy with poor prognosis. Further work is needed to determine the optimal multimodal treatment regimen.


Subject(s)
Carcinoma, Adenosquamous , Carcinoma, Pancreatic Ductal , Delivery of Health Care, Integrated , Pancreatic Neoplasms , Humans , Retrospective Studies , Carcinoma, Adenosquamous/pathology , Carcinoma, Adenosquamous/surgery , Pancreatectomy , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms
4.
J Geriatr Oncol ; 13(7): 952-961, 2022 09.
Article in English | MEDLINE | ID: mdl-35597730

ABSTRACT

INTRODUCTION: Pancreatic ductal adenocarcinoma (PDAC) mainly occurs in older adults. Since randomized clinical trials (RCTs) provide the highest-quality evidence incorporated in NCCN recommendations, the underrepresentation of older patients in RCTs challenges guidelines' external validity and limits the solidity of evidence in this specific population. MATERIALS AND METHODS: The study aimed to investigate external validity of NCCN guidelines for PDCA and the impact of reference studies eligibility on overall survival (OS) in a real-world older population. We retrieved RCTs supporting NCCN recommendations for management of PDAC and identified ten topics. We matched a cohort of 707 PDAC patients aged ≥70 years from the Moffitt Cancer Center database with eligibility criteria of 96 reference RCTs to check the proportion of patients eligible for at least two RCTs. Eligibility >60% was rated full validity, 30%-60% partial validity and < 30% limited validity. We also performed log-rank test to assessed whether "eligibility" status affects OS, stratifying by age (70-74; 75-79; ≥80). RESULTS AND DISCUSSION: We found full validity for neoadjuvant (57/73 patients; 69.86%), locally advanced (28/39; 71.79%) and second line (88/110; 80%) treatment, while lowest validity was found for adjuvant chemotherapy (37/86; 43%). Eligible status was correlated with a significant OS benefit for adjuvant chemoradiation (p = 0.002) in all-comers and for first-line polychemotherapy in patients aged ≥80 (p = 0.01). Our analysis supports the limitation of guidelines' external validity in older patients, and hints at possible correlations with survival, although no definitive conclusions can be drawn at this stage. Renewing RCT design with broader eligibility criteria might help increase inclusion of older and thus strengthen the evidence.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/therapy , Chemoradiotherapy, Adjuvant , Humans , Neoadjuvant Therapy , Pancreatic Neoplasms
5.
Expert Rev Anticancer Ther ; 22(2): 155-167, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34989653

ABSTRACT

INTRODUCTION: Pancreatic Ductal Adenocarcinoma (PDAC) is an aggressive disease with poor outcomes. One of the reasons for the dismal prognosis resides in its impressive ability to alter the nutritional status of patients who develop malnutrition, cachexia, anorexia, and sarcopenia in most cases. The ideal way to measure such changes in PDAC patients, in order to readily identify them and avoid complications or discontinuations of treatment is a relatively unexplored area. In addition, most PDAC patients experience pancreatic exocrine insufficiency (PEI) that contributes to the complex puzzle of malnutrition and that can be treated with Pancreatic Enzyme Replacement Therapy (PERT). AREAS COVERED: We review current knowledge on the impact of nutritional status on both surgical and medical treatments for PDAC, reporting available data on the causes of malnutrition, characteristics, and advantages of different tools to investigate nutritional status and possible strategies to improve patient outcomes. EXPERT OPINION: All PDAC patients should receive a careful nutritional assessment at diagnosis, and this should be repeated alongside their treatment path. Screening tools and biochemical variables or scores are associated with prognosis, but bioimpedance vector analysis (BIVA) and radiological assessment of body composition seem more accurate in predicting clinical outcomes and postoperative complications.


Subject(s)
Carcinoma, Pancreatic Ductal , Exocrine Pancreatic Insufficiency , Malnutrition , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Exocrine Pancreatic Insufficiency/complications , Exocrine Pancreatic Insufficiency/diagnosis , Humans , Malnutrition/complications , Malnutrition/therapy , Nutritional Status , Pancreas/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms
6.
Pancreas ; 51(8): 923-929, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36607936

ABSTRACT

ABSTRACT: Pancreatic ductal adenocarcinoma (PDAC) is currently an increasing contributor to cancer-related mortality. Despite advances in cancer treatment, PDAC survival rates have remained roughly unchanged over the years. Specifically, late diagnosis and insensitivity to currently available therapeutic regimens have been identified as the main causes for its poor survival. Pancreatic exocrine insufficiency (PEI) is a typical complication associated with PDAC diagnosis and pancreatic surgery. Pancreatic exocrine insufficiency, a major contributor to maldigestion in PDAC, is often not treated because it remains undetected because of lack of overt signs and symptoms. In this review, we will focus on the major consequences of PEI, including the inadequacy of lipase excretion, which results in deficiency of fat-soluble vitamins. Because PDAC is known for its immune-high jacking mechanisms, we describe key features in which deficiencies of fat-soluble vitamins may contribute to the aggressive biological behavior and immune evasion in PDAC. Because PEI has been shown to worsen survival rates in patients with PDAC, detecting PEI and the related fat-soluble vitamin deficits at the time of PDAC diagnosis is critical. Moreover, timely supplementation of pancreatic enzymes and fat-soluble vitamins may improve outcomes for PDAC patients.


Subject(s)
Avitaminosis , Carcinoma, Pancreatic Ductal , Exocrine Pancreatic Insufficiency , Pancreatic Neoplasms , Humans , Vitamins/therapeutic use , Exocrine Pancreatic Insufficiency/etiology , Exocrine Pancreatic Insufficiency/complications , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/complications , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/complications , Immune System , Avitaminosis/complications , Pancreatic Neoplasms
7.
Pathol Oncol Res ; 27: 1610048, 2021.
Article in English | MEDLINE | ID: mdl-34955688

ABSTRACT

The poor prognosis of pancreatic ductal adenocarcinoma (PDAC) is frequently associated to high treatment resistance. Gemcitabine (GEM) alone or in combination is the most used chemotherapy for unresecable PDACs. Here we studied whether modulated electro-hyperthermia (mEHT), a non-invasive complementary treatment, can support the effect of GEM on PDAC cells in vitro. The LD20 for the GEM-resistant Panc1 cells proved to be 200× higher than for the drug-sensitive Capan1. The mEHT alone caused significant apoptosis in Capan1 cultures as confirmed by the elevated SubG1 phase cell fraction and increased number of cleaved Caspase-3 positive cells 48 h after treatment, with an additive effect when GEM was used after hyperthermia. These were accompanied by reduced number of G1, S, and G2/M phase cells and elevated expression of the cyclin-dependent kinase inhibitor p21waf1 protein. In GEM-resistant Panc1 cells, an initial apoptosis was detected by flow cytometry 24 h after mEHT ± GEM treatment, which however diminished by 48 h at persistent number of cleaved Caspase-3 positive tumor cells. Though GEM monotherapy reduced the number of tumor progenitor colonies in Capan1 cell line, an additive colony inhibitory effect of mEHT was observed after mEHT + GEM treatment. The heat shock induced Hsp27 and Hsp70 proteins, which are known to sensitize PDAC cells to GEM were upregulated in both Capan1 and Panc1 cells 24 h after mEHT treatment. The level of E-Cadherin, a cell adhesion molecule, increased in Capan1 cells after mEHT + GEM treatment. In conclusion, in GEM-sensitive PDAC cells mEHT treatment alone induced cell death and cell cycle inhibition and improved GEM efficiency in combination, which effects were milder and short-term up to 24 h in the GEM-resistant Panc1 cells. Our data further support the inclusion of hyperthermia, in particular of mEHT, into the traditional oncotherapy regimens of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Hyperthermia, Induced , Pancreatic Neoplasms/therapy , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Adhesion/drug effects , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Combined Modality Therapy , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Heat-Shock Proteins/metabolism , Humans , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Gemcitabine
8.
Surg Today ; 51(11): 1872-1876, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34086127

ABSTRACT

Active hexose-correlated compound (AHCC) is a standardized extract from cultured Lentinula edodes mycelia, used as a potent biological response modifier in cancer treatment. We evaluated the nutritional effect of AHCC, given during neoadjuvant therapy, to patients with pancreatic ductal adenocarcinoma (PDAC). Thirty patients with resectable or borderline-resectable PDAC received neoadjuvant therapy with gemcitabine plus S-1. We compared, retrospectively, the outcomes of 15 patients who received AHCC combined with neoadjuvant therapy with those of 15 patients who did not receive AHCC combined with neoadjuvant therapy. The median changes of the neutrophil-to-lymphocyte ratio (NLR) and prognostic nutrition index (PNI) were significantly better in the AHCC group. The relative dose intensity of neoadjuvant therapy was also significantly higher in the AHCC group. Thus, AHCC may improve the nutritional status during neoadjuvant therapy of patients with pancreatic ductal adenocarcinoma. To validate these results and examine the long-term impact of AHCC, a prospective phase II study for PDAC is ongoing.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/physiopathology , Carcinoma, Pancreatic Ductal/therapy , Deoxycytidine/analogs & derivatives , Neoadjuvant Therapy , Nutrition Assessment , Nutrition Therapy , Nutritional Status , Oxonic Acid/administration & dosage , Pancreatic Neoplasms/physiopathology , Pancreatic Neoplasms/therapy , Phytotherapy , Polysaccharides/administration & dosage , Tegafur/administration & dosage , Aged , Aged, 80 and over , Deoxycytidine/administration & dosage , Drug Combinations , Female , Humans , Male , Middle Aged , Polysaccharides/isolation & purification , Shiitake Mushrooms/chemistry , Treatment Outcome , Gemcitabine
9.
J Surg Oncol ; 123(6): 1449-1459, 2021 May.
Article in English | MEDLINE | ID: mdl-33831249

ABSTRACT

Despite overall advances in cancer therapy, patients with pancreatic ductal adenocarcinoma continue to have a poor prognosis. While adjuvant therapy is still considered standard, there is mounting evidence that neoadjuvant therapy confers similar benefits in patients with locally advanced disease. The primary measures of response are radiographic, biochemical, margin status, and pathologic. Given overall low response rates and the need for new treatment strategies, standard metrics remain important to the investigation of new systemic agents.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Pancreatic Neoplasms/therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/surgery , Fluorouracil/administration & dosage , Humans , Irinotecan/administration & dosage , Leucovorin/administration & dosage , Neoadjuvant Therapy , Oxaliplatin/administration & dosage , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/surgery
10.
PLoS One ; 16(3): e0248633, 2021.
Article in English | MEDLINE | ID: mdl-33735191

ABSTRACT

Factors for overall survival after pancreatic ductal adenocarcinoma (PDAC) seem to be nodal status, chemotherapy administration, UICC staging, and resection margin. However, there is no consensus on the definition for tumor free resection margin. Therefore, univariate OS as well as multivariate long-term survival using cancer center data was analyzed with regards to two different resection margin definitions. Ninety-five patients met inclusion criteria (pancreatic head PDAC, R0/R1, no 30 days mortality). OS was analyzed in univariate analysis with respect to R-status, CRM (circumferential resection margin; positive: ≤1mm; negative: >1mm), nodal status, and chemotherapy administration. Long-term survival >36 months was modelled using multivariate logistic regression instead of Cox regression because the distribution function of the dependent data violated the requirements for the application of this test. Significant differences in OS were found regarding the R status (Median OS and 95%CI for R0: 29.8 months, 22.3-37.4; R1: 15.9 months, 9.2-22.7; p = 0.005), nodal status (pN0 = 34.7, 10.4-59.0; pN1 = 17.1, 11.5-22.8; p = 0.003), and chemotherapy (with CTx: 26.7, 20.4-33.0; without CTx: 9.7, 5.2-14.1; p < .001). OS according to CRM status differed on a clinically relevant level by about 12 months (CRM positive: 17.2 months, 11.5-23.0; CRM negative: 29.8 months, 18.6-41.1; p = 0.126). A multivariate model containing chemotherapy, nodal status, and CRM explained long-term survival (p = 0.008; correct prediction >70%). Chemotherapy, nodal status and resection margin according to UICC R status are univariate factors for OS after PDAC. In contrast, long-term survival seems to depend on wider resection margins than those used in UICC R classification. Therefore, standardized histopathological reporting (including resection margin size) should be agreed upon.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Pancreas/pathology , Pancreatic Neoplasms/therapy , Pancreaticoduodenectomy/statistics & numerical data , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Chemotherapy, Adjuvant/methods , Chemotherapy, Adjuvant/statistics & numerical data , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Irinotecan/administration & dosage , Kaplan-Meier Estimate , Leucovorin/administration & dosage , Male , Margins of Excision , Middle Aged , Neoadjuvant Therapy/methods , Neoadjuvant Therapy/statistics & numerical data , Neoplasm Staging , Oxaliplatin/administration & dosage , Pancreas/surgery , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreaticoduodenectomy/standards , Retrospective Studies , Risk Assessment/methods , Risk Assessment/standards , Risk Assessment/statistics & numerical data , Time Factors , Treatment Outcome
11.
Surg Oncol ; 37: 101542, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33740629

ABSTRACT

BACKGROUND: Present treatment strategy for unresectable locally advanced (UR-LA) pancreatic ductal adenocarcinoma (PDAC) patients is controversial. Hence, a triple-modal therapy, which is a multidisciplinary strategy, was designed for patients with UR-LA PDAC by adding hyperthermia to conventional chemoradiotherapy at our institution. In this study we aimed to evaluate the effectiveness of this strategy. METHODS: Data of 21 UR-LA PDAC patients who underwent the triple-modal treatment were retrospectively analyzed for evaluating the safety and oncological effect of the treatment. The treatment schedule included, five concurrent infusions of gemcitabine (800 mg/m2) followed by hyperthermia (1 h) and X-ray (2 Gy) or proton beam radiation (2.7 Gy) on days 1, 8, 15, 29, and 36. Additional radiotherapies applied a total dose of 50 Gy/25 fr for X-ray radiation or 67.5 Gy/25 fr for proton beam radiation. RESULTS: Median overall survival (OS) was 23.6 months. Conversion surgery was performed in 5 patients (23.8%), and a R0 margin could be achieved in 4 of them; however, their median OS (16.3 months) tended to be shorter than that of the patients who did not undergo resection (23.6 months, p = 0.562). Further, the median OS of patients who underwent proton beam radiation (28.0 months) was significantly longer than that of patients who underwent X-ray radiation (13.9 months, p = 0.045). Most adverse events were manageable, except for one grade 3 gastric ulcer. The median tumor size and marker reduction rates were -17% and -91%, respectively. The tumor responses were partial response, stable disease, and progressive disease in 3, 15, and 3 patients, respectively. CONCLUSION: Triple-modal strategy, especially when combined with proton beam radiation, is feasible and results in favorable survival outcomes in patients with UR-LA PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/therapy , Chemoradiotherapy , Hyperthermia, Induced , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/therapy , Adult , Aged , Combined Modality Therapy , Female , Humans , Male , Middle Aged , Retrospective Studies , Survival Rate
12.
J Surg Res ; 257: 605-615, 2021 01.
Article in English | MEDLINE | ID: mdl-32947122

ABSTRACT

BACKGROUND: The clinicopathologic factors associated with the survival of patients with pancreatic ductal adenocarcinoma (PDAC) during the different phases of neoadjuvant treatment (NT)-at diagnosis, restaging, or postoperatively-remain unclear. METHODS: Data of patients with PDAC who underwent pancreatic resection after NT between 2008 and 2018 were retrospectively collected. Clinicopathologic characteristics and outcomes were compared stratified by resection margin status. Three multivariable regression models (at diagnosis, restaging, and postoperatively) were constructed to assess the temporal impact of different prognostic factors on all-cause survival (ACS) and disease-free survival (DFS). RESULTS: All patients were diagnosed with a nonmetastatic PDAC and were appropriate candidates for NT according to the current National Comprehensive Cancer Network guidelines. From a total of 83 patients, 57 (68.7%) had a negative resection margin >1 mm (R0), whereas 26 patients (31.3%) had a positive resection margin (R1). At diagnosis, planned procedure (P = 0.017) and CA19-9 >100 U/mL (P = 0.047) were independent prognostic factors of decreased ACS. At restaging, planned procedure (P = 0.017), FOLFIRINOX (P = 0.026), and tumor size >30 mm (P = 0.030) were independent prognostic factors for increased and decreased ACS, respectively. Postoperatively, R0 was an independent prognostic factor for improved ACS (P = 0.005) and DFS (P = 0.002), whereas adjuvant therapy (P = 0.006) was associated with increased ACS. Lymph node involvement (P = 0.019) was associated with decreased DFS. CONCLUSIONS: At diagnosis, restaging, and postoperatively, different, relevant clinicopathologic factors significantly impact the survival of patients with nonmetastatic PDAC undergoing NT. An R0 resection remains the most important prognostic factor and therefore should be the primary goal of surgical treatment in the neoadjuvant setting.


Subject(s)
Carcinoma, Pancreatic Ductal/mortality , Neoadjuvant Therapy , Pancreatectomy , Pancreatic Neoplasms/mortality , Aged , Boston/epidemiology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Female , Humans , Male , Middle Aged , Pancreas/pathology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Prognosis , Proportional Hazards Models , Retrospective Studies
13.
J Am Coll Surg ; 232(4): 405-413, 2021 04.
Article in English | MEDLINE | ID: mdl-33338577

ABSTRACT

BACKGROUND: Historically, a positive margin after pancreatectomy for pancreatic ductal adenocarcinoma (PDAC) was associated with decreased survival. In an era when neoadjuvant chemotherapy (NAC) is being used frequently, the prognostic significance of margin status is unclear. STUDY DESIGN: Patients with localized PDAC who received NAC and underwent pancreatectomy from 2011 to 2018 were identified from a single-institution database. Patients with fewer than 2 months of NAC, R2 resection, or fewer than 90 days of follow-up were excluded. A positive margin included tumors within 1 mm of the surgical margin. RESULTS: Four hundred and sixty-eight patients met inclusion criteria. Median age was 65 years and 53% were female. Preoperative clinical staging demonstrated that most had locally advanced (n = 222 [47%]) or borderline resectable (n = 172 [37%]) disease. Median follow-up was 18.5 months (interquartile range 10.6 to 30.0 months). Median duration of NAC was 119 days (interquartile range 87 to 168 days). FOLFIRINOX was first-line therapy for 67%, and 73% received neoadjuvant radiotherapy. Most underwent pancreaticoduodenectomy (69%). Forty percent were node-positive and 80% had an R0 resection. Fifty-six percent received at least 1 cycle of adjuvant therapy. Median overall survival and recurrence-free survival were 22.0 months (95% CI, 19.4 to 25.1 months) and 11.0 months (95% CI, 10.0 to 12.1 months). On multivariate analysis, margin status was not a significant predictor of overall survival or recurrence-free survival. Factors associated with overall survival included clinical stage, duration of NAC, nodal status, histopathologic treatment response score, and receipt of adjuvant chemotherapy. CONCLUSIONS: Microscopic margin positivity is not associated with recurrence and survival in localized PDAC patients resected after treatment with NAC. Aggressive surgical extirpation in high-volume centers should be considered in selected patients after extensive NAC.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Neoadjuvant Therapy/methods , Neoplasm Recurrence, Local/epidemiology , Pancreatic Neoplasms/therapy , Pancreaticoduodenectomy/statistics & numerical data , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Chemotherapy, Adjuvant/methods , Disease-Free Survival , Female , Fluorouracil/therapeutic use , Follow-Up Studies , Humans , Irinotecan/therapeutic use , Leucovorin/therapeutic use , Male , Margins of Excision , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Neoplasm Staging , Oxaliplatin/therapeutic use , Pancreas/pathology , Pancreas/surgery , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prospective Studies , Retrospective Studies
14.
Ann Surg ; 272(3): 481-486, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32740235

ABSTRACT

OBJECTIVE: The optimal neoadjuvant therapy for resectable pancreatic ductal adenocarcinoma (PDA) and the impact on surgical outcomes remains unclear. METHODS: S1505 (NCT02562716) was a randomized phase II study of perioperative chemotherapy with mFOLFIRINOX (Arm 1) or gemcitabine/nab-paclitaxel (Arm 2). Measured parameters included resection rate, margin positivity, pathologic response, and toxicity. RESULTS: Between 2015 and 2018, 147 patients were randomized. Of these, 44 (30%) were deemed ineligible (43 by central review). Of the 103 eligible patients, 77 (76%) completed preoperative therapy and underwent surgery; reasons patients did not undergo surgery included toxicity related to preoperative therapy (n = 9), progression (n = 9), or other (n = 7). Of the 77, 73 (95%) underwent successful resection; 21 (29%) required vascular reconstruction, 62 (85%) had negative (R0) margins, and 24 (33%) had a complete or major pathologic response to therapy. The grade 3-5 postoperative complication rate was 16%. Of the 73 patients completing surgery, 57 (78%) started and 46 (63%) completed postoperative therapy. This study represents the first prospective trial evaluating modern systemic therapy delivered in a neoadjuvant/perioperative format for resectable PDA. CONCLUSIONS: We have demonstrated: (1) Based on the high percentage of enrolled, but ineligible patients, it is clear that adherence to strict definitions of resectable PDA is challenging; (2) Patients can tolerate modern systemic therapy and undergo successful surgical resection without prohibitive perioperative complications; (3) Completion of adjuvant therapy in the perioperative format is difficult; (4) Major pathologic response rate of 33% is encouraging.


Subject(s)
Albumins/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Deoxycytidine/analogs & derivatives , Paclitaxel/therapeutic use , Pancreatectomy , Pancreatic Neoplasms/therapy , Perioperative Care/methods , Aged , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/diagnosis , Combined Modality Therapy , Deoxycytidine/therapeutic use , Female , Fluorouracil/therapeutic use , Follow-Up Studies , Humans , Immunosuppressive Agents/therapeutic use , Irinotecan/therapeutic use , Leucovorin/therapeutic use , Male , Middle Aged , Neoplasm Staging , Oxaliplatin/therapeutic use , Pancreatic Neoplasms/diagnosis , Prospective Studies , Treatment Outcome , Gemcitabine
15.
Clin Cancer Res ; 26(18): 5007-5018, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32611647

ABSTRACT

PURPOSE: To evaluate the prognostic value of posttreatment fibrosis in human PDAC patients, and to compare a type I collagen targeted MRI probe, CM-101, to the standard contrast agent, Gd-DOTA, for their abilities to identify FOLFIRINOX-induced fibrosis in a murine model of PDAC. EXPERIMENTAL DESIGN: Ninety-three chemoradiation-treated human PDAC samples were stained for fibrosis and outcomes evaluated. For imaging, C57BL/6 and FVB mice were orthotopically implanted with PDAC cells and FOLFIRINOX was administered. Mice were imaged with Gd-DOTA and CM-101. RESULTS: In humans, post-chemoradiation PDAC tumor fibrosis was associated with longer overall survival (OS) and disease-free survival (DFS) on multivariable analysis (OS P = 0.028, DFS P = 0.047). CPA increased the prognostic accuracy of a multivariable logistic regression model comprised of previously established PDAC risk factors [AUC CPA (-) = 0.76, AUC CPA (+) = 0.82]. In multiple murine orthotopic PDAC models, FOLFIRINOX therapy reduced tumor weight (P < 0.05) and increased tumor fibrosis by collagen staining (P < 0.05). CM-101 MR signal was significantly increased in fibrotic tumor regions. CM-101 signal retention was also increased in the more fibrotic FOLFIRINOX-treated tumors compared with untreated controls (P = 0.027), consistent with selective probe binding to collagen. No treatment-related differences were observed with Gd-DOTA imaging. CONCLUSIONS: In humans, post-chemoradiation tumor fibrosis is associated with OS and DFS. In mice, our MR findings indicate that translation of collagen molecular MRI with CM-101 to humans might provide a novel imaging technique to monitor fibrotic response to therapy to assist with prognostication and disease management.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Chemoradiotherapy, Adjuvant , Neoadjuvant Therapy/methods , Neoplasm Recurrence, Local/epidemiology , Pancreas/pathology , Pancreatic Neoplasms/therapy , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Collagen/analysis , Collagen/metabolism , Disease-Free Survival , Female , Fibrosis , Fluorouracil/administration & dosage , Follow-Up Studies , Heterocyclic Compounds/administration & dosage , Humans , Irinotecan/administration & dosage , Leucovorin/administration & dosage , Magnetic Resonance Imaging/methods , Male , Mice , Middle Aged , Molecular Imaging/methods , Molecular Probes/administration & dosage , Neoplasm Recurrence, Local/prevention & control , Organometallic Compounds/administration & dosage , Oxaliplatin/administration & dosage , Pancreas/diagnostic imaging , Pancreas/surgery , Pancreatectomy , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Retrospective Studies , Survival Analysis , Xenograft Model Antitumor Assays
16.
Oncology (Williston Park) ; 34(6): 196-201, 2020 Jun 10.
Article in English | MEDLINE | ID: mdl-32609864

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a disease marked by high rates of mortality; it is mostly incurable at the time of diagnosis. Only about 7% of patients survive 5 years after diagnosis. Diagnosis at a late stage and rapid progression with minimal response to available treatments are the main reasons for this poor outcome. It is crucial to identify individuals at high risk of developing PDAC so preventive and early detection measures can be employed. Approximately 10% to 15% of PDAC cases have a hereditary or familial basis. In the majority of PDAC cases, no main causative gene has been identified, but several known germline pathogenic mutations have been shown to be related to an increased risk of this cancer. The presence of 2 or more patients with pancreatic cancer within the circle of first-degree relatives, without the presence of a causative germline mutation, is defined as familial pancreatic cancer; this accounts for 4% to 10% of PDAC. Based on the growing evidence supporting the benefit of germline genetic testing in patients with PDAC, both the American Society of Clinical Oncology and the National Comprehensive Cancer Network recently updated their guidelines to include recommendations around genetic testing for patients with pancreatic cancer. However, there is no general consensus on the group of patients and individuals who should be studied and screened. We present a demonstrative case and review the available data on hereditary and familial PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma/genetics , Germ-Line Mutation , Pancreatic Neoplasms/genetics , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/diagnostic imaging , Carcinoma/therapy , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/therapy , Genetic Testing/methods , Humans , Male , Neoplastic Syndromes, Hereditary/diagnostic imaging , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Pancreatectomy/methods , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/therapy , Risk Factors
17.
Biosci Rep ; 40(7)2020 07 31.
Article in English | MEDLINE | ID: mdl-32701143

ABSTRACT

BACKGROUND: Hypermethylation of gene promoters plays an important role in tumorigenesis. The present study aimed to identify and validate promoter methylation-driven genes (PMDGs) for pancreatic ductal adenocarcinoma (PDAC). METHODS: Based on GSE49149 and the PDAC cohort of The Cancer Genome Atlas (TCGA), differential analyses of promoter methylation, correlation analysis, and Cox regression analysis were performed to identify PMDGs. The promoter methylation level was assessed by bisulfite sequencing polymerase chain reaction (BSP) in paired tumor and normal tissues of 72 PDAC patients. Kaplan-Meier survival analyses were performed to evaluate the clinical value of PMDGs. RESULTS: In GSE49149, the ß-value of the dipeptidyl peptidase like 6 (DPP6) promoter was significantly higher in tumor compared with normal samples (0.50 vs. 0.24, P<0.001). In the PDAC cohort of TCGA, the methylation level of the DPP6 promoter was negatively correlated with mRNA expression (r = -0.54, P<0.001). In a multivariate Cox regression analysis, hypermethylation of the DPP6 promoter was an independent risk factor for PDAC (hazard ratio (HR) = 543.91, P=0.002). The results of BSP revealed that the number of methylated CG sites in the DPP6 promoter was greater in tumor samples than in normal samples (7.43 vs. 2.78, P<0.001). The methylation level of the DPP6 promoter was moderately effective at distinguishing tumor from normal samples (area under ROC curve (AUC) = 0.74, P<0.001). Hypermethylation of the DPP6 promoter was associated with poor overall (HR = 3.61, P<0.001) and disease-free (HR = 2.01, P=0.016) survivals for PDAC patients. CONCLUSION: These results indicate that DPP6 promoter methylation is a potential prognostic biomarker for PDAC.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/mortality , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Nerve Tissue Proteins/genetics , Pancreatic Neoplasms/mortality , Potassium Channels/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Chemotherapy, Adjuvant , CpG Islands/genetics , DNA Methylation , Disease-Free Survival , Epigenesis, Genetic , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Middle Aged , Pancreas/pathology , Pancreas/surgery , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Pancreaticoduodenectomy , Prognosis , Promoter Regions, Genetic/genetics , Radiotherapy, Adjuvant
18.
Int J Mol Sci ; 21(14)2020 Jul 19.
Article in English | MEDLINE | ID: mdl-32707717

ABSTRACT

The poor outcome of pancreas ductal adenocarcinomas (PDAC) is frequently linked to therapy resistance. Modulated electro-hyperthermia (mEHT) generated by 13.56 MHz capacitive radiofrequency can induce direct tumor damage and promote chemo- and radiotherapy. Here, we tested the effect of mEHT either alone or in combination with radiotherapy using an in vivo model of Panc1, a KRAS and TP53 mutant, radioresistant PDAC cell line. A single mEHT shot of 60 min induced ~50% loss of viable cells and morphological signs of apoptosis including chromatin condensation, nuclear shrinkage and apoptotic bodies. Most mEHT treatment related effects exceeded those of radiotherapy, and these were further amplified after combining the two modalities. Treatment related apoptosis was confirmed by a significantly elevated number of annexin V single-positive and cleaved/activated caspase-3 positive tumor cells, as well as sub-G1-phase tumor cell fractions. mEHT and mEHT+radioterapy caused the moderate accumulation of γH2AX positive nuclear foci, indicating DNA double-strand breaks and upregulation of the cyclin dependent kinase inhibitor p21waf1 besides the downregulation of Akt signaling. A clonogenic assay revealed that both mono- and combined treatments affected the tumor progenitor/stem cell populations too. In conclusion, mEHT treatment can contribute to tumor growth inhibition and apoptosis induction and resolve radioresistance of Panc1 PDAC cells.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Hyperthermia, Induced/methods , Pancreatic Neoplasms/therapy , Apoptosis , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Combined Modality Therapy , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Humans , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Radiation Tolerance , Radiofrequency Therapy
19.
Biochim Biophys Acta Rev Cancer ; 1874(1): 188387, 2020 08.
Article in English | MEDLINE | ID: mdl-32579889

ABSTRACT

Late detection, compromised immune system, and chemotherapy resistance underlie the poor patient prognosis for pancreatic ductal adenocarcinoma (PDAC) patients, making it the 3rd leading cause of cancer-related deaths in the United States. Cooperation between the tumor cells and the immune system leads to the immune escape and eventual establishment of the tumor. For more than 20 years, sincere efforts have been made to intercept the tumor-immune crosstalk and identify the probable therapeutic targets for breaking self-tolerance toward tumor antigens. However, the success of these studies depends on detailed examination and understanding of tumor-immune cell interactions, not only in the primary tumor but also at distant systemic niches. Innate and adaptive arms of the immune system sculpt tumor immunogenicity, where they not only aid in providing an amenable environment for their survival but also act as a driver for tumor relapse at primary or distant organ sites. This review article highlights the key events associated with tumor-immune communication and associated immunosuppression at both local and systemic microenvironments in PDAC. Furthermore, we discuss the approaches and benefits of targeting both local and systemic immunosuppression for PDAC patients. The present articles integrate data from clinical and genetic mouse model studies to provide a widespread consensus on the role of local and systemic immunosuppression in undermining the anti-tumor immune responses against PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Immunotherapy/methods , Pancreatic Neoplasms/therapy , Tumor Escape/drug effects , Tumor Microenvironment/immunology , Adaptive Immunity/drug effects , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/drug effects , Bone Marrow/immunology , Bone Marrow/pathology , Cancer Vaccines/administration & dosage , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Chemotherapy, Adjuvant/methods , Clinical Trials as Topic , Combined Modality Therapy/methods , Disease Models, Animal , Disease-Free Survival , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Immunity, Innate/drug effects , Irinotecan/pharmacology , Irinotecan/therapeutic use , Leucovorin/pharmacology , Leucovorin/therapeutic use , Lymph Node Excision , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/surgery , Mice , Mice, Transgenic , Neoadjuvant Therapy/methods , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Pancreas/immunology , Pancreas/pathology , Pancreas/surgery , Pancreatectomy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Spleen/immunology , Spleen/pathology , Spleen/surgery , Splenectomy , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Transplantation, Autologous/methods
20.
Clin Nutr ; 39(11): 3385-3394, 2020 11.
Article in English | MEDLINE | ID: mdl-32184025

ABSTRACT

BACKGROUND & AIMS: Nutritional impairments are highly frequent in pancreatic cancer even in the early stages and have a significant impact on outcomes. The aim of this prospective study was to investigate immune and nutritional impairments, their interrelations and impacts on outcomes in an unselected cohort of patients scheduled for pancreatoduodenectomy due to suspicion of pancreatic cancer. METHODS: All consecutive patients scheduled for pancreatoduodenectomy at Vilnius University Hospital Santaros Klinikos between January 2016 and November 2018 were recruited into the study according to the inclusion/exclusion criteria. Patients were randomly allocated into the groups of nutritional intervention with immunonutrition vs. control and stratified into the groups of pancreatic ductal adenocarcinoma (PDAC) vs. other pancreatic tumors. Nutritional evaluation included screening (NRS 2002), anthropometric measurements, bioelectrical impedance analysis and lumbar skeletal muscle index (LSMI). Inflammatory indicators were measured before and after surgery. Surgical outcomes were assessed 30 days postoperatively using Comprehensive Complication Index (CCI). RESULTS: Although increased nutritional risk was identified in 22.4% of patients, 41.4% were finally diagnosed with cachexia. While cachexia was predominantly diagnosed in underweight patients, sarcopenia was detected across all BMI categories and 11.7% of obese patients had sarcopenia. Decreased LSMI was identified in 52.5% of patients as compared to decreased phase angle in 39% of patients and decreased fat free mass index in only 3.4% of patients. Regression model indicate a large effect of nutritional indicators on CCI (R2 coefficient 71.1%). In comparison to patients with other pancreatic tumors, patients with PDAC had a characteristic pattern of increased systemic inflammation prior to surgery and decreased inflammation postoperatively (p = 0.02). CONCLUSIONS: A high rate of nutritional impairments was identified in our cohort of patients with early pancreatic cancer, including abnormal body composition phenotypes. They produced negative effects on postoperative outcomes. The highest diagnostic rates were obtained with LSMI measurement, while the highest value for prognostication was attained with the inclusion of multiple objective nutritional state indicators.


Subject(s)
Carcinoma, Pancreatic Ductal/physiopathology , Nutritional Status , Pancreatic Neoplasms/physiopathology , Pancreaticoduodenectomy , Postoperative Complications/etiology , Aged , Anthropometry , Body Mass Index , Cachexia/epidemiology , Cachexia/etiology , Carcinoma, Pancreatic Ductal/complications , Carcinoma, Pancreatic Ductal/therapy , Dietary Supplements , Electric Impedance , Female , Humans , Lumbar Vertebrae/physiopathology , Male , Middle Aged , Nutrition Assessment , Nutrition Therapy/methods , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/therapy , Perioperative Care/methods , Preoperative Period , Prospective Studies , Regression Analysis , Sarcopenia/epidemiology , Sarcopenia/etiology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL