Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Surg Oncol ; 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39155666

ABSTRACT

BACKGROUND: Chemotherapy enhances survival rates for pancreatic cancer (PC) patients postsurgery, yet less than 60% complete adjuvant therapy, with a smaller fraction undergoing neoadjuvant treatment. Our study aimed to predict which patients would complete pre- or postoperative chemotherapy through machine learning (ML). METHODS: Patients with resectable PC identified in our institutional pancreas database were grouped into two categories: those who completed all intended treatments (i.e., surgery plus either neoadjuvant or adjuvant chemotherapy), and those who did not. We applied logistic regression with lasso penalization and an extreme gradient boosting model for prediction, and further examined it through bootstrapping for sensitivity. RESULTS: Among 208 patients, the median age was 69, with 49.5% female and 62% white participants. Most had an Eastern Cooperative Oncology Group (ECOG) performance status of ≤2. The PC predominantly affected the pancreatic head. Neoadjuvant and adjuvant chemotherapies were received by 26% and 47.1%, respectively, but only 49% completed all treatments. Incomplete therapy was correlated with older age and lower ECOG status. Negative prognostic factors included worsening diabetes, age, congestive heart failure, high body mass index, family history of PC, initial bilirubin levels, and tumor location in the pancreatic head. The models also flagged other factors, such as jaundice and specific cancer markers, impacting treatment completion. The predictive accuracy (area under the receiver operating characteristic curve) was 0.67 for both models, with performance expected to improve with larger datasets. CONCLUSIONS: Our findings underscore the potential of ML to forecast PC treatment completion, highlighting the importance of specific preoperative factors. Increasing data volumes may enhance predictive accuracy, offering valuable insights for personalized patient strategies.

2.
Cureus ; 16(3): e56934, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38665761

ABSTRACT

Advanced pancreatic cancer is one of the prominent contributors to cancer-related mortality globally. Chemotherapy, especially gemcitabine, is generally used for the treatment of advanced pancreatic cancer. Despite the treatment, the fatality rate for advanced pancreatic cancer is alarmingly high. Thus, the dire need for better treatment alternatives has drawn focus to cancer vaccinations. The Wilms tumor gene (WT1), typically associated with Wilms tumor, is found to be excessively expressed in some cancers, such as pancreatic cancer. This characteristic feature is harvested to develop cancer vaccines against WT1. This review aims to systematically summarize the clinical trials investigating the efficacy and safety of WT1 vaccines in patients with advanced pancreatic cancer. An extensive literature search was conducted on databases Medline, Web of Science, ScienceDirect, and Google Scholar using the keywords "Advanced pancreatic cancer," "Cancer vaccines," "WT1 vaccines," and "Pulsed DC vaccines," and the results were exclusively studied to construct this review. WT1 vaccines work by introducing peptides from the WT1 protein to trigger an immune response involving cytotoxic T lymphocytes via antigen-presenting cells. Upon activation, these lymphocytes induce apoptosis in cancer cells by specifically targeting those with increased WT1 levels. WT1 vaccinations, which are usually given in addition to chemotherapy, have demonstrated clinically positive results and minimal side effects. However, there are several challenges to their widespread use, such as the immunosuppressive nature of tumors and heterogeneity in expression. Despite these limitations, the risk-benefit profile of cancer vaccines is encouraging, especially for the WT1 vaccine in the treatment of advanced pancreatic cancer. Considering the fledgling status of their development, large multicentric, variables-matched, extensive analysis across diverse demographics is considered essential.

3.
Adv Mater ; 36(25): e2401017, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38573785

ABSTRACT

Glycolysis-dominant metabolic pathway in cancer cells can promote their therapeutic resistance against radiotherapy (RT). Carbon monoxide (CO) as a glycolysis inhibitor can enhance the efficiency of RT. Herein, an X-ray responsive CO-releasing nanocomposite (HA@AuNC@CO) based on strong host-guest interactions between the radiosensitizer and CO donor for enhanced RT is developed. The encapsulated gold nanoclusters (CD-AuNCs) can effectively generate cytotoxic reactive oxygen species (ROS) under X-ray radiation, which not only directly inactivate cancer cells but also induce in situ CO gas generation from adamantane modified metal carbonyl (Ada-CO) for glycolysis inhibition. Both in vitro and in vivo results demonstrate that HA@AuNC@CO exhibits active targeting toward CD44 overexpressed cancer cells, along with excellent inhibition of glycolysis and efficient RT against cancer. This study offers a new strategy for the combination of gas therapy and RT in tumor treatment.


Subject(s)
Carbon Monoxide , Glycolysis , Gold , Metal Nanoparticles , Reactive Oxygen Species , Gold/chemistry , Carbon Monoxide/chemistry , Humans , Animals , Metal Nanoparticles/chemistry , Reactive Oxygen Species/metabolism , Cell Line, Tumor , X-Rays , Glycolysis/drug effects , Mice , Neoplasms/drug therapy , Neoplasms/therapy , Neoplasms/metabolism , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacology
4.
PeerJ ; 12: e16805, 2024.
Article in English | MEDLINE | ID: mdl-38250721

ABSTRACT

Background: Pancreatic cancer is a highly aggressive and fatal disease with limited treatment options and poor prognosis for patients. This study aimed to investigate the impact of XYA-2 {N-(3,7-dimethyl-2,6-octadienyl)-2-aza-2-deoxychaetoviridin A}, a nitrogenated azaphilon previously reported from a deep-sea-derived fungus on the progression of pancreatic cancer cells. Methods: The inhibitory effects of XYA-2 on cell proliferation, clonogenic potential, cell cycle progression, apoptosis, migration, and invasion were assessed using various assays. The CCK-8 assay, clone formation assay, flow cytometry assay, wound healing assay, and transwell assay were employed to evaluate cell proliferation, clonogenic potential, cell cycle progression, apoptosis, migration, and invasion, respectively. Moreover, we employed RNA-seq and bioinformatics analyses to uncover the underlying mechanism by which XYA-2 influences pancreatic cancer cells. The revealed mechanism was subsequently validated through qRT-PCR. Results: Our results demonstrated that XYA-2 dose-dependently inhibited the proliferation of pancreatic cancer cells and induced cell cycle arrest and apoptosis. Additionally, XYA-2 exerted a significant inhibitory effect on the invasion and migration of cancer cells. Moreover, XYA-2 was found to regulate the expression of genes involved in multiple cancer-related pathways based on our RNA-seq and bioinformatics analysis. Conclusion: These findings highlight the potential of XYA-2 as a promising therapeutic option for the treatment of pancreatic cancer.


Subject(s)
Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/drug therapy , Apoptosis , Pancreas , Cell Proliferation , Pancreatic Hormones , Signal Transduction
5.
Therap Adv Gastroenterol ; 17: 17562848231224943, 2024.
Article in English | MEDLINE | ID: mdl-38250014

ABSTRACT

Background: The impact of KRAS mutation testing on pancreatic ductal adenocarcinoma (PDAC) samples by endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) for reducing the need to repeat EUS-FNA has been demonstrated. Such testing however is not part of standard practice for endoscopic ultrasound-guided fine-needle biopsy (EUS-FNB). Objectives: We aim to analyse the proportion of non-contributive samples by EUS-FNB and to evaluate the impact of KRAS mutation testing on the diagnosis, theranostics and survival. Design: In this retrospective study, the impact on diagnosis and survival of KRAS testing for contributive and non-contributive samples by EUS-FNB was analysed. Methods: The EUS-FNB samples, combined with KRAS testing using the Idylla® technique on liquid-based cytology from patients with PDAC between February 2019 and May 2023, were retrospectively reviewed. The cytology results were classified according to the guidelines of the World Health Organization System for Reporting Pancreaticobiliary Cytopathology (WHOSRPC). Results: A total of 85 EUS-FNB specimens were reviewed. In all, 25 EUS-FNB samples did not lead to a formal diagnosis of PDAC according to the WHOSRPC (30.2%). Out of these 25, 11 (44%) could have been considered positive for a PDAC diagnosis thanks to the KRAS mutation test without carrying out further diagnosis procedures. The sensitivity of KRAS mutation testing using the Idylla technique was 98.6%. According to the available data, survival rates were not statistically different depending on the type of mutation. Conclusion: KRAS mutation testing on liquid-based cytology using the Idylla or equivalent technique, combined with the PDAC EUS-FNB sample, should become a standard for diagnosis to avoid delaying treatment by doing another biopsy. Furthermore, knowledge of the KRAS status from treatment initiation could be used to isolate mutations requiring targeted treatments or inclusion in clinical research trials, especially for wild-type KRAS PDAC.


Diagnostic and theranostic interest of searching for a KRAS mutation in echoendoscopic ultrasound biopsies of pancreatic adenocarcinomas The echoendoscopic ultrasound diagnostic of pancreatic adenocarcinomas sometimes remains difficult due to the nature of these tumors with a particular microenvironment. For more than 30 years, several authors have underlined the importance of searching for a KRAS mutation on samples taken by echoendoscopic ultrasound to improve diagnostic performance. However, this research is not common practice. Our retrospective study made it possible to review the files of 85 patients with pancreatic adenocarcinoma in whom an echoendoscopic ultrasound biopsy was performed with a search for the KRAS mutation (with second-generation fine needle biopsy). Forty-four percent could have been considered positive for the diagnosis of PDAC thanks to the search for the KRAS mutation without repeating new samples. Furthermore, knowledge of the KRAS mutation type from diagnosis would make it possible to isolate mutations justifying possible targeted treatments.

6.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1038507

ABSTRACT

Objective To calculate the absorbed dose of 90Y TheraSphere in the pancreas and the surrounding sensitive organs after the administration in the treatment of pancreatic cancer through the establishment of an individual voxel model, and to provide technical support for the clinical application of 90Y TheraSphere in the treatment of pancreatic cancer. Methods An individualized voxel model was constructed in Geant4 software based on the CT images of the patient. 12 monoenergetic electron specific absorption fractions (SAFs) in the range of 0.01 to 1 MeV were calculated and validated against the ICRP data. The model and method were used to calculate the absorbed doses in the target organs under uniform and nonuniform distribution of 90Y microspheres in the pancreas. Results The relative errors between the SAF values calculated based on the individualized voxel model and the ICRP data after mass calibration were less than 3.89%. When 90Y was uniformly distributed in the pancreas, the absorbed dose in the pancreas was 4.69 × 10−7 Gy/Bq; the absorbed doses in the liver, kidneys, and spleen were 6.15 × 10−12, 6 × 10−12, and 1.65 × 10−11 Gy/Bq, respectively. When 90Y was distributed within the tumor, the absorbed dose in the tumor was 6.69 × 10−6 Gy/Bq and the absorbed dose in normal pancreas was 5.72 × 10−8 Gy/Bq. The fitted relationship between tumor volume V and administered activity A at the prescribed dose of 120 Gy was quadratic, with relatively low activity required for concentrated administration in the center of the tumor. Conclusion The Monte Carlo dose calculation method based on individual voxel model accurately predicted the absorbed doses in the surrounding sensitive organs (liver, kidneys, and spleen) when 90Y TheraSphere was used to treat pancreatic cancer. These results and the analysis of the factors affecting the drug delivery activity will provide data support for the clinical research of 90Y TheraSphere in pancreatic cancer.

7.
Hematol Oncol Clin North Am ; 36(5): 1011-1018, 2022 10.
Article in English | MEDLINE | ID: mdl-36154782

ABSTRACT

Advanced pancreatic cancer remains one of the deadliest malignancies in 2022. Although there has been significant progress in treatment options with improved outcomes in many cancers, this growth has been slow in pancreatic cancer. This article examines specific components of approved first- and second-line therapies for advanced pancreatic cancer treatment and their effectiveness and concludes with a brief exploration of future directions for targeted therapies.


Subject(s)
Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Pancreatic Neoplasms
8.
BMC Cancer ; 22(1): 925, 2022 Aug 27.
Article in English | MEDLINE | ID: mdl-36030217

ABSTRACT

BACKGROUND: The gap in treatment and health outcomes after diagnosis of pancreatic cancer is a major public health concern. We aimed to investigate the differences in the health outcomes and treatment of pancreatic cancer patients in healthcare vulnerable and non-vulnerable areas. METHODS: This retrospective cohort study evaluated data from the Korea National Health Insurance Corporation-National Sample Cohort from 2002 to 2019. The position value for relative comparison index was used to define healthcare vulnerable areas. Cox proportional hazard regression was used to estimate the risk of mortality in pancreatic cancer patients according to healthcare vulnerable areas, and multiple logistic regression was used to estimate the difference in treatment. RESULTS: Among 1,975 patients, 279 (14.1%) and 1,696 (85.9%) lived in the healthcare vulnerable and non-vulnerable areas, respectively. Compared with the non-vulnerable area, pancreatic cancer patients in the vulnerable area had a higher risk of death at 3 months (hazard ratio [HR]: 1.33, 95% confidence interval [CI] = 1.06-1.67) and 6 months (HR: 1.23, 95% CI = 1.03-1.48). In addition, patients with pancreatic cancer in the vulnerable area were less likely to receive treatment than patients in the non-vulnerable area (odds ratio [OR]: 0.70, 95% CI = 0.52-0.94). This trend was further emphasized for chemotherapy (OR: 0.68, 95% CI = 0.48-0.95). CONCLUSION: Patients with pancreatic cancer belonging to medically disadvantaged areas receive less treatment and have a higher risk of death. This may be a result of the late diagnosis of pancreatic cancer among these patients.


Subject(s)
Pancreatic Neoplasms , Cohort Studies , Healthcare Disparities , Humans , Republic of Korea , Retrospective Studies , Pancreatic Neoplasms
9.
World J Gastroenterol ; 27(35): 5851-5889, 2021 Sep 21.
Article in English | MEDLINE | ID: mdl-34629806

ABSTRACT

Pancreatic cancer is one of the solid tumors with the worst prognosis. Five-year survival rate is less than 10%. Surgical resection is the only potentially curative treatment, but the tumor is often diagnosed at an advanced stage of the disease and surgery could be performed in a very limited number of patients. Moreover, surgery is still associated with high post-operative morbidity, while other therapies still offer very disappointing results. This article reviews every aspect of pancreatic cancer, focusing on the elements that can improve prognosis. It was written with the aim of describing everything you need to know in 2021 in order to face this difficult challenge.


Subject(s)
Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/therapy , Prognosis
10.
Biomedicines ; 9(4)2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33917380

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer deaths in the US, and it is expected to be the second leading cause of cancer deaths by 2030. The lack of effective early screening tests and alarming symptoms with early undetectable micro-metastasis at the time of presentation play a vital role in the high death rate from pancreatic cancer. In addition to this, the low mutation burden in pancreatic cancer, low immunological profile, dense tumorigenesis stroma, and decreased tumor sensitivity to cytotoxic drugs contribute to the low survival rates in PDAC patients. Despite breakthroughs in chemotherapeutic and immunotherapeutic drugs, pancreatic cancer remains one of the solid tumors that exhibit meager curative rates. Therefore, researchers must dedicate more effort to understanding the pathology and immunological behavior of PDAC, in addition to properly utilizing more advanced screening modalities and new therapeutic agents. In our review, we focus mainly on the latest updates from clinical guidelines and novel therapies that have been recently investigated or are under investigation for PDAC. We used PubMed as a search tool for finding original research articles addressing the latest developments in diagnosing and treating PDAC. Additionally, we also used the clinical trials published on clinicaltrialsgov as sources for our data.

11.
Cell Mol Gastroenterol Hepatol ; 11(1): 161-179, 2021.
Article in English | MEDLINE | ID: mdl-32810598

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) is resistant to most therapeutics owing to dense fibrotic stroma orchestrated by cancer-associated pancreatic stellate cells (CAPaSC). CAPaSC also support cancer cell growth, metastasis, and resistance to apoptosis. Currently, there is no effective therapy for PDAC that specifically targets CAPaSC. We previously reported a rationally designed protein, ProAgio, that targets integrin αvß3 at a novel site and induces apoptosis in integrin αvß3-expressing cells. Because both CAPaSC and angiogenic endothelial cells express high levels of integrin αvß3, we aimed to analyze the effects of ProAgio in PDAC tumor. METHODS: Expression of integrin αvß3 was examined in both patient tissue and cultured cells. The effects of ProAgio on CAPaSC were analyzed using an apoptosis assay kit. The effects of ProAgio in PDAC tumor were studied in 3 murine tumor models: subcutaneous xenograft, genetic engineered (KrasG12D; p53R172H; Pdx1-Cre, GEM-KPC) mice, and an orthotopic KrasG12D; p53R172H; Pdx1-Cre (KPC) model. RESULTS: ProAgio induces apoptosis in CAPaSC. ProAgio treatment significantly prolonged survival of a genetically engineered mouse-KPC and orthotopic KPC mice alone or in combination with gemcitabine (Gem). ProAgio specifically induced apoptosis in CAPaSC, resorbed collagen, and opened collapsed tumor vessels without an increase in angiogenesis in PDAC tumor, enabling drug delivery into the tumor. ProAgio decreased intratumoral insulin-like growth factor 1 levels as a result of depletion of CAPaSC and consequently decreased cytidine deaminase, a Gem metabolism enzyme in cancer cells, and thereby reduced resistance to Gem-induced apoptosis. CONCLUSIONS: Our study suggests that ProAgio is an effective PDAC treatment agent because it specifically depletes CAPaSC and eliminates tumor angiogenesis, thereby enhancing drug delivery and Gem efficacy in PDAC tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Integrin alphaVbeta3/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Pancreatic Stellate Cells/drug effects , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Disease Models, Animal , Humans , Integrin alphaVbeta3/analysis , Integrin alphaVbeta3/metabolism , Mice , Mice, Transgenic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Primary Cell Culture , Xenograft Model Antitumor Assays
12.
Anticancer Res ; 40(4): 1789-1796, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32234867

ABSTRACT

Pancreatic cancer is often diagnosed due to the patient seeking medical attention for abdominal pain. It is among the most painful cancers, with pain severity strongly correlating with prognosis. Perineural invasion is a prominent feature of pancreatic cancer and often the first route of metastasis resulting in neuropathic pain. While surgical pain is present, it is generally short-lived; chemo- and radio-therapy associated side effect pain is often longer lasting and more difficult to manage. Treatment-induced mucositis in response to chemotherapy occurs throughout the GI tract resulting in infection-prone ulcers on the lip, buccal mucosa, palate or tongue. Cisplatin treatment is associated with axonal neuropathy in the dorsal root ganglion, although other large sensory fibers can be affected. Opioid-induced hyperalgesia can also emerge in patients. Along with traditional means to address pain, neurolytic celiac plexus block of afferent nociceptive fibers has been reported to be effective in 74% of patients. Moreover, as cancer treatments become more effective and result in improved survival, treatment-related side effects become more prevalent. Here, pancreatic cancer and treatment associated pain are reviewed along with current treatment strategies. Potential future therapeutic strategies to target the pathophysiology underlying pancreatic cancer and pain induction are also presented.


Subject(s)
Cancer Pain/drug therapy , Mucositis/drug therapy , Pain Management/adverse effects , Pancreatic Neoplasms/drug therapy , Abdominal Pain/drug therapy , Abdominal Pain/etiology , Analgesics, Opioid/adverse effects , Analgesics, Opioid/therapeutic use , Bupivacaine/therapeutic use , Cancer Pain/pathology , Cisplatin/adverse effects , Cisplatin/therapeutic use , Humans , Mucositis/chemically induced , Pain Measurement , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/pathology
13.
World J Gastroenterol ; 24(43): 4846-4861, 2018 Nov 21.
Article in English | MEDLINE | ID: mdl-30487695

ABSTRACT

This review aims to outline the most up-to-date knowledge of pancreatic adenocarcinoma risk, diagnostics, treatment and outcomes, while identifying gaps that aim to stimulate further research in this understudied malignancy. Pancreatic adenocarcinoma is a lethal condition with a rising incidence, predicted to become the second leading cause of cancer death in some regions. It often presents at an advanced stage, which contributes to poor five-year survival rates of 2%-9%, ranking firmly last amongst all cancer sites in terms of prognostic outcomes for patients. Better understanding of the risk factors and symptoms associated with this disease is essential to inform both health professionals and the general population of potential preventive and/or early detection measures. The identification of high-risk patients who could benefit from screening to detect pre-malignant conditions such as pancreatic intraepithelial neoplasia, intraductal papillary mucinous neoplasms and mucinous cystic neoplasms is urgently required, however an acceptable screening test has yet to be identified. The management of pancreatic adenocarcinoma is evolving, with the introduction of new surgical techniques and medical therapies such as laparoscopic techniques and neo-adjuvant chemoradiotherapy, however this has only led to modest improvements in outcomes. The identification of novel biomarkers is desirable to move towards a precision medicine era, where pancreatic cancer therapy can be tailored to the individual patient, while unnecessary treatments that have negative consequences on quality of life could be prevented for others. Research efforts must also focus on the development of new agents and delivery systems. Overall, considerable progress is required to reduce the burden associated with pancreatic cancer. Recent, renewed efforts to fund large consortia and research into pancreatic adenocarcinoma are welcomed, but further streams will be necessary to facilitate the momentum needed to bring breakthroughs seen for other cancer sites.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/therapy , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/therapy , Precancerous Conditions/diagnosis , Biomarkers, Tumor/analysis , Carcinoma, Pancreatic Ductal/epidemiology , Carcinoma, Pancreatic Ductal/pathology , Chemoradiotherapy, Adjuvant/methods , Early Detection of Cancer/methods , Humans , Incidence , Mass Screening/methods , Neoadjuvant Therapy/methods , Pancreas/pathology , Pancreas/surgery , Pancreatectomy/methods , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/pathology , Precancerous Conditions/epidemiology , Precancerous Conditions/pathology , Prognosis , Quality of Life , Risk Factors , Survival Rate , Treatment Outcome
14.
Gastroenterology ; 155(1): 29-32, 2018 07.
Article in English | MEDLINE | ID: mdl-29567081

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is resistant to T-cell-mediated immunotherapy. We engineered T cells to transiently express a messenger RNA encoding a chimeric antigen receptor (CAR) specific for mesothelin, a protein that is overexpressed by PDAC cells. We performed a phase I study to evaluate the safety and efficacy of adoptive cell therapy with autologous mesothelin-specific CAR T cells (CARTmeso cells) in 6 patients with chemotherapy-refractory metastatic PDAC. Patients were given intravenous CARTmeso cells 3 times weekly for 3 weeks. None of the patients developed cytokine release syndrome or neurologic symptoms and there were no dose-limiting toxicities. Disease stabilized in 2 patients, with progression-free survival times of 3.8 and 5.4 months. We used 18F-2-fluoro-2-deoxy-D-glucose (FDG)-positron emission tomography/computed tomography imaging to monitor the metabolic active volume (MAV) of individual tumor lesions. The total MAV remained stable in 3 patients and decreased by 69.2% in 1 patient with biopsy-proven mesothelin expression; in this patient, all liver lesions had a complete reduction in FDG uptake at 1 month compared with baseline, although there was no effect on the primary PDAC. Transient CAR expression was detected in patients' blood after infusion and led to expansion of new immunoglobulin G proteins. Our results provide evidence for the potential antitumor activity of messenger RNA CARTmeso cells, as well as PDAC resistance to the immune response.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , GPI-Linked Proteins/immunology , Immunotherapy, Adoptive/methods , Pancreatic Neoplasms/drug therapy , RNA, Messenger/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/transplantation , Aged , Carcinoma, Pancreatic Ductal/secondary , Disease-Free Survival , Female , Fluorodeoxyglucose F18 , Humans , Male , Mesothelin , Middle Aged , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals , Survival Rate , T-Lymphocytes/immunology , Transplantation, Autologous
15.
Oncol Lett ; 15(2): 1441-1448, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29434835

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has one of the poorest prognoses of all malignancy types. To improve the survival of patients with PDAC, the development of novel anticancer agents is warranted. Riproximin (Rpx) is a newly identified plant lectin, which was isolated from Ximenia americana. The ribosome inactivating protein of type II exhibits potent anticancer activity as recently demonstrated. The rat PDAC cell line ASML was used for in vitro and in vivo studies. The antiproliferative effect of Rpx was assessed using an MTT assay. The modulation of proteins involved in apoptosis was evaluated using western blotting. Tumor-bearing nude rats were treated with Rpx, gemcitabine (GEM) or dinaline (DIN) as single agents, or a combination of Rpx with GEM, or DIN. Rpx was administered intraperitoneally at doses of 1.7-5.4 µg/kg, three times/week, GEM was administered intravenously (50 mg/kg/week) and DIN perorally (10 mg/kg, 5 times/week). Rpx inhibited ASML cell proliferation at IC50-values of 0.8-172 pM, caused apoptosis and reduced tumor growth significantly by 90% (P<0.05). The survival rate of rats was significantly increased (21.8 days for Rpx treated vs. 17.6 days for control rats; P=0.05). Higher doses of Rpx caused no further reduction in tumor size when compared with the low dose of Rpx or a combination of Rpx with GEM, or DIN. The standard drug GEM alone was less effective compared with Rpx. In addition, DIN was ineffective, and in combination, reduced the activity of Rpx. These results suggest that Rpx has an evident potential for use in pancreatic cancer treatment. Further experiments are required in order to elucidate its affinity for certain cancer cells and to optimize the combination therapy with other antineoplastic agents.

16.
Expert Rev Anticancer Ther ; 18(3): 295-304, 2018 03.
Article in English | MEDLINE | ID: mdl-29334794

ABSTRACT

INTRODUCTION: Pancreatic cancer is known to be the deadliest of all common cancers. Despite all efforts in pancreatic cancer treatment, the five-year survival rates at diagnosis over the past 20 years have only increased from 5% to 8%. Assuming that pancreatic cancer is going to become the second most frequent cause of cancer related death in the next 20 years, we are all encouraged to treat patients in clinical trials to gain improvements in this devastating disease. Areas covered: This review will provide a summary of pancreatic cancer treatment over the last 20 years, starting with the pivotal study in 1997 which showed the superiority of gemcitabine over 5-FU in advanced pancreatic cancer and is marked as the beginning of a new era in pancreatic cancer treatment. This review will also focus on improvements in different areas of treatment, including pancreatic surgery, adjuvant treatment, neoadjuvant therapy and palliative therapy. Expert commentary: The treatment of pancreatic cancer has changed substantially in the last 20 years compared to almost no improvements in the decades before. This provides hope that more effective treatment options will become available in the near future. Particularly, new concepts such as neoadjuvant therapy in resectable and borderline-resectable tumors may potentially shift treatment strategies.


Subject(s)
Antineoplastic Agents/therapeutic use , Palliative Care/methods , Pancreatic Neoplasms/therapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Fluorouracil/therapeutic use , Humans , Neoadjuvant Therapy/methods , Neoadjuvant Therapy/trends , Palliative Care/trends , Pancreatic Neoplasms/pathology , Survival Rate , Gemcitabine
17.
J Gastrointest Cancer ; 49(1): 41-49, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28005258

ABSTRACT

INTRODUCTION: In the US, gastroenterologists (GIs) often inform patients of the initial diagnosis of pancreatic cancer. Thus, GIs are frequently the first physicians to provide vital information regarding treatment strategies and options to patients which can have significant impact on subsequent clinical decision-making. Since treatments for pancreatic cancer are rapidly evolving, it may be challenging for GIs to maintain an adequate knowledge base required to provide accurate cursory information or avoid providing inaccurate data to patients at a very sensitive point in time in their care. However, little to no published data are available on the treatment awareness, knowledge, and comfort of GIs in the United States who diagnose pancreatic cancer. METHODS: This study evaluated the self-reported management of pancreatic cancer, including patient discussion, familiarity with treatments, and use of guidelines. A survey was developed and fielded to US academic and community GIs in 2013. In 2015, the survey was redistributed as a follow-up to determine whether familiarity and practice has shifted; two questions were added, all other items remained identical. For the 2013 sample, 432 GIs were contacted and 113 valid responses were collected (26.2). For the 2015 sample, 712 GIs were contacted and 126 valid responses were collected (17.7%). Analysis compared differences between academic and community gastroenterologists and gastroenterologists performing endoscopic ultrasound (EUS) versus those that do not. RESULTS: Self-reported familiarity with pancreatic cancer therapies has not significantly increased from 2013 to 2015, as gastroenterologists report highest familiarity with 5-fluorouracil and gemcitabine. In the 2015 sample, 68% of academic and 58% of community gastroenterologists entered gemcitabine when identifying therapies FDA-approved for treatment of pancreatic cancer. However, 16% of academic and 24% of community gastroenterologists indicated that they were unaware of which therapies are approved, and some indicated therapies that are not FDA-approved for the treatment of pancreatic cancer, such as capecitabine (10%) and paclitaxel (7%). Gastroenterologists in 2015 are significantly more likely than in 2013 to discuss clinical trial enrollment with their patients with metastatic pancreatic cancer (5.5 on a 10-point scale vs 4.2, P = .013) but were not more confident in their ability to conduct such discussions. When managing patients with pancreatic cancer, academic, and community gastroenterologists responded that they were most likely to refer to guidelines developed by their professional organizations, such as the AGA and ACG. However, these groups have not developed specific guidelines for the management of patients with pancreatic cancer. CONCLUSIONS: As gastroenterologists are frequently the first physicians to disclose a diagnosis of pancreatic cancer, education is needed to improve familiarity with current available treatments, clinical trials, and emerging therapies and resources to advise their patients.


Subject(s)
Gastroenterologists/statistics & numerical data , Pancreatic Neoplasms/therapy , Practice Patterns, Physicians'/statistics & numerical data , Female , Humans , Immunohistochemistry , Male , Middle Aged , Pancreatic Neoplasms/pathology , Retrospective Studies , Surveys and Questionnaires , United States
18.
World J Gastroenterol ; 22(5): 1779-86, 2016 Feb 07.
Article in English | MEDLINE | ID: mdl-26855537

ABSTRACT

Endoscopic ultrasonography (EUS) is a technique with an established role in the diagnosis and staging of gastro-intestinal tumors. In recent years, the spread of new devices dedicated to tissue sampling has improved the diagnostic accuracy of EUS fine-needle aspiration. The development of EUS-guided drainage of the bilio-pancreatic region and abdominal fluid collections has allowed EUS to evolve into an interventional tool that can replace more invasive procedures. Emerging techniques applying EUS in pancreatic cancer treatment and in celiac neurolysis have been described. Recently, confocal laser endomicroscopy has been applied to EUS as a promising technique for the in vivo histological diagnosis of gastro-intestinal, bilio-pancreatic and lymph node lesions. In this state-of-the-art review, we report the most recent data from the literature regarding EUS devices, interventional EUS, EUS-guided confocal laser endomicroscopy and EUS pancreatic cancer treatment, and we also provide an overview of their principles, clinical applications and limitations.


Subject(s)
Drainage/methods , Endosonography/methods , Gastrointestinal Diseases/diagnostic imaging , Gastrointestinal Diseases/therapy , Gastrointestinal Tract/diagnostic imaging , Endoscopic Ultrasound-Guided Fine Needle Aspiration , Gastrointestinal Diseases/pathology , Gastrointestinal Tract/pathology , Humans , Microscopy, Confocal , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/therapy , Predictive Value of Tests , Prognosis
19.
Gastroenterology ; 149(1): 201-10, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25888329

ABSTRACT

BACKGROUND & AIMS: Immunotherapies that induce T-cell responses have shown efficacy against some solid malignancies in patients and mice, but these have little effect on pancreatic ductal adenocarcinoma (PDAC). We investigated whether the ability of PDAC to evade T-cell responses induced by immunotherapies results from the low level of immunogenicity of tumor cells, the tumor's immunosuppressive mechanisms, or both. METHODS: Kras(G12D/+);Trp53(R172H/+);Pdx-1-Cre (KPC) mice, which develop spontaneous PDAC, or their littermates (controls) were given subcutaneous injections of a syngeneic KPC-derived PDAC cell line. Mice were then given gemcitabine and an agonist of CD40 to induce tumor-specific immunity mediated by T cells. Some mice were also given clodronate-encapsulated liposomes to deplete macrophages. Tumor growth was monitored. Tumor and spleen tissues were collected and analyzed by histology, flow cytometry, and immunohistochemistry. RESULTS: Gemcitabine in combination with a CD40 agonist induced T-cell-dependent regression of subcutaneous PDAC in KPC and control mice. In KPC mice given gemcitabine and a CD40 agonist, CD4(+) and CD8(+) T cells infiltrated subcutaneous tumors, but only CD4(+) T cells infiltrated spontaneous pancreatic tumors (not CD8(+) T cells). In mice depleted of Ly6C(low) F4/80(+) extratumoral macrophages, the combination of gemcitabine and a CD40 agonist stimulated infiltration of spontaneous tumors by CD8(+) T cells and induced tumor regression, mediated by CD8(+) T cells. CONCLUSIONS: Ly6C(low) F4/80(+) macrophages that reside outside of the tumor microenvironment regulate infiltration of T cells into PDAC and establish a site of immune privilege. Strategies to reverse the immune privilege of PDAC, which is regulated by extratumoral macrophages, might increase the efficacy of T-cell immunotherapy for patients with PDAC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/drug therapy , Immunotherapy/methods , Macrophages/cytology , Macrophages/immunology , Pancreatic Neoplasms/drug therapy , Animals , Antimetabolites, Antineoplastic/pharmacology , CD40 Antigens/agonists , CD8-Positive T-Lymphocytes/drug effects , Carcinoma, Pancreatic Ductal/immunology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Immunohistochemistry , Macrophages/drug effects , Mice , Pancreatic Neoplasms/immunology , Gemcitabine , Pancreatic Neoplasms
20.
J Gastrointest Oncol ; 5(4): 280-95, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25083302

ABSTRACT

Although pancreatic adenocarcinoma is the fourth leading cause of cancer death, only modest improvement has been observed in the past two decades, single agent gemcitabine has been the only standard treatment in patients with advanced disease. Recently newer agents such as nab-paclitaxel, nimotuzumab and regimens such as FOLFIRINOX have been shown to have promising activity being superior to gemcitabine as a single agent. With better understanding of tumour biology coupled with the improvements in targeted and immunotherapies, there is increasing expectation for better response rates and extended survival in pancreatic cancer.

SELECTION OF CITATIONS
SEARCH DETAIL