Pub Date : 2020-07-01Epub Date: 2020-07-22DOI: 10.21037/apc.2020.03.03
Mengni He, MacKenzie Henderson, Stephen Muth, Adrian Murphy, Lei Zheng
Pancreatic ductal adenocarcinoma (PDAC) is in urgent need of better diagnostic and therapeutic methods due to its late diagnosis, limited treatment options and poor prognosis. Finding the right animal models to recapitulate the tumor molecular pathogenesis and tumor microenvironment (TME) complexity is critical for preclinical immunotherapeutic and non-immunotherapeutic treatment developments. In this review, we summarize and evaluate popular preclinical animal models including patient-derived xenograft models, humanized mouse models, genetically engineered mouse models, and syngeneic mouse models. Through comparisons between these models in different research settings, we hope to provide guidance in finding the most relevant preclinical models to suit various research purposes.
{"title":"Preclinical mouse models for immunotherapeutic and non-immunotherapeutic drug development for pancreatic ductal adenocarcinoma.","authors":"Mengni He, MacKenzie Henderson, Stephen Muth, Adrian Murphy, Lei Zheng","doi":"10.21037/apc.2020.03.03","DOIUrl":"https://doi.org/10.21037/apc.2020.03.03","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is in urgent need of better diagnostic and therapeutic methods due to its late diagnosis, limited treatment options and poor prognosis. Finding the right animal models to recapitulate the tumor molecular pathogenesis and tumor microenvironment (TME) complexity is critical for preclinical immunotherapeutic and non-immunotherapeutic treatment developments. In this review, we summarize and evaluate popular preclinical animal models including patient-derived xenograft models, humanized mouse models, genetically engineered mouse models, and syngeneic mouse models. Through comparisons between these models in different research settings, we hope to provide guidance in finding the most relevant preclinical models to suit various research purposes.</p>","PeriodicalId":8372,"journal":{"name":"Annals of Pancreatic Cancer","volume":"3 ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2020-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.21037/apc.2020.03.03","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38300963","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2020-05-01Epub Date: 2020-05-20DOI: 10.21037/apc-2020-pda-05
Arnav Mehta, William L Hwang, Colin Weekes
Metastatic pancreatic adenocarcinoma remains one of the deadliest cancer diagnoses with 5-year survival rates as low as 3%. For decades, gemcitabine remained the mainstay of systemic therapy before the approvals of FOLFIRINOX and gemcitabine with nab-paclitaxel. Despite these advances in the early 2010s, almost all patients progress on systemic chemotherapy and significant effort is needed to identify novel therapeutic targets. A promising array of approaches is currently under investigation, enabled by deeper understanding of the immune system within the tumor microenvironment (TME) and of the key vulnerabilities in pathways essential for tumor survival. In this review, we will explore the different approaches to boost tumor immunity and to target tumor metabolic pathways that are currently under clinical investigation for systemic treatment, and highlight the promising therapeutic areas that may give rise to the next generation of therapies for pancreatic cancer.
{"title":"The present and future of systemic and microenvironment-targeted therapy for pancreatic adenocarcinoma.","authors":"Arnav Mehta, William L Hwang, Colin Weekes","doi":"10.21037/apc-2020-pda-05","DOIUrl":"10.21037/apc-2020-pda-05","url":null,"abstract":"<p><p>Metastatic pancreatic adenocarcinoma remains one of the deadliest cancer diagnoses with 5-year survival rates as low as 3%. For decades, gemcitabine remained the mainstay of systemic therapy before the approvals of FOLFIRINOX and gemcitabine with nab-paclitaxel. Despite these advances in the early 2010s, almost all patients progress on systemic chemotherapy and significant effort is needed to identify novel therapeutic targets. A promising array of approaches is currently under investigation, enabled by deeper understanding of the immune system within the tumor microenvironment (TME) and of the key vulnerabilities in pathways essential for tumor survival. In this review, we will explore the different approaches to boost tumor immunity and to target tumor metabolic pathways that are currently under clinical investigation for systemic treatment, and highlight the promising therapeutic areas that may give rise to the next generation of therapies for pancreatic cancer.</p>","PeriodicalId":8372,"journal":{"name":"Annals of Pancreatic Cancer","volume":"3 ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2020-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/21/b6/nihms-1650265.PMC7720884.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38693227","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2020-03-01Epub Date: 2020-03-09DOI: 10.21037/apc.2020.02.01
Alejandro Recio-Boiles, Jessica Vondrak, Summana Veeravelli, James J Mancuso, Kathylynn Saboda, Denise J Roe, Irbaz Bin Riaz, Aaron J Scott, Emad Elquza, Ali McBride, Hani M Babiker
Background: Only 15-20% of pancreatic ductal adenocarcinoma (PDAC) patients are upfront surgical candidates at presentation, and for this cohort of patients, the 5-year survival is a mere 20% despite adjuvant therapy. Previous data indicate that in clinical practice most of these cases are "borderline-resectable," and there is currently no mature data on perioperative treatment.
Methods: We performed a retrospective electronic chart review of patients with "borderline-resectable"PDAC treated at an academic comprehensive cancer center, dividing them into groups based on surgery alone, surgery plus neoadjuvant, adjuvant, or neoadjuvant plus adjuvant perioperative treatment groups. The objectives were to determine the median overall survival (mOS), progression-free survival (PFS) and disease-free survival (DFS). Statistical analysis was performed to assess the association of demographic, tumor traits, and interventions with OS, PFS and DFS.
Results: Only surgery followed by adjuvant therapy showed an increase in mOS [hazard ratio (HR) 0.22; 95% CI, 0.09-0.51; P<0.001), after adjustment for radiation (yes vs. no), resection margins (R0 vs. R1 or R2), and tumor location (head vs. body or tail). Patients who received adjuvant therapy after surgery had 2.1 times greater odds to be alive at 24 months after diagnosis than those who had surgery alone (P=0.015). PFS and DFS were not statistically significantly different among treatment groups after adjustment. Those whose disease was located in the head of the pancreas had a significantly improved OS (HR =0.27; 95% CI, 0.11-0.64; P=0.003), PFS (HR =0.40; 95% CI, 0.17-0.94; P=0.035), and DFS (HR =0.30; 95% CI, 0.13-0.67; P=0.004). Negative margins led to a significant improvement in PFS (HR =0.30; 95% CI, 0.16-0.57; P<0.001) and DFS (HR =0.30; 95% CI, 0.16-0.57; P<0.001). Those who received radiation had a non-significantly improved OS, PFS, and DFS (P>0.05).
Conclusions: Our study corroborated that patients treated with adjuvant therapy after surgical resection had an mOS benefit as reported on prior phase III clinical trials. Patients with "borderline-resectable" pancreatic cancer are encouraged to participate in a clinical trial or clinically be treated with adjuvant therapy until more mature results from the ongoing perioperative prospective study are available.
背景:只有15%-20%的胰腺导管腺癌(PDAC)患者在就诊时适合先期手术治疗,尽管进行了辅助治疗,但这部分患者的5年生存率仅为20%。以往的数据表明,在临床实践中,这些病例中的大多数都是 "边缘可切除",目前还没有关于围手术期治疗的成熟数据:方法:我们对在一家学术综合癌症中心接受治疗的 "边缘可切除 "PDAC 患者进行了回顾性电子病历审查,根据单纯手术组、手术加新辅助治疗组、辅助治疗组或新辅助治疗加辅助治疗围手术期治疗组将患者分为几组。目的是确定中位总生存期(mOS)、无进展生存期(PFS)和无病生存期(DFS)。统计分析评估了人口统计学、肿瘤特征和干预措施与OS、PFS和DFS的关系:结果:只有手术后接受辅助治疗的患者的mOS[危险比(HR)0.22;95% CI,0.09-0.51;Pvs.no]、切除边缘(R0 vs. R1或R2)和肿瘤位置(头部 vs. 体部或尾部)均有所增加。术后接受辅助治疗的患者在确诊后24个月内存活的几率是单纯手术患者的2.1倍(P=0.015)。经调整后,各治疗组的 PFS 和 DFS 在统计学上无明显差异。病变位于胰腺头部的患者的OS(HR=0.27;95% CI,0.11-0.64;P=0.003)、PFS(HR=0.40;95% CI,0.17-0.94;P=0.035)和DFS(HR=0.30;95% CI,0.13-0.67;P=0.004)均有明显改善。阴性边缘可显著改善PFS(HR=0.30;95% CI,0.16-0.57;P0.05):我们的研究证实,正如之前的III期临床试验所报告的那样,手术切除后接受辅助治疗的患者可获得mOS益处。我们鼓励 "边缘可切除 "胰腺癌患者参加临床试验或接受辅助治疗,直到正在进行的围手术期前瞻性研究得出更成熟的结果。
{"title":"Analyzing outcomes of neoadjuvant and adjuvant treatment for borderline-resectable pancreatic adenocarcinoma in the perioperative period at an academic institution.","authors":"Alejandro Recio-Boiles, Jessica Vondrak, Summana Veeravelli, James J Mancuso, Kathylynn Saboda, Denise J Roe, Irbaz Bin Riaz, Aaron J Scott, Emad Elquza, Ali McBride, Hani M Babiker","doi":"10.21037/apc.2020.02.01","DOIUrl":"10.21037/apc.2020.02.01","url":null,"abstract":"<p><strong>Background: </strong>Only 15-20% of pancreatic ductal adenocarcinoma (PDAC) patients are upfront surgical candidates at presentation, and for this cohort of patients, the 5-year survival is a mere 20% despite adjuvant therapy. Previous data indicate that in clinical practice most of these cases are \"borderline-resectable,\" and there is currently no mature data on perioperative treatment.</p><p><strong>Methods: </strong>We performed a retrospective electronic chart review of patients with \"borderline-resectable\"PDAC treated at an academic comprehensive cancer center, dividing them into groups based on surgery alone, surgery plus neoadjuvant, adjuvant, or neoadjuvant plus adjuvant perioperative treatment groups. The objectives were to determine the median overall survival (mOS), progression-free survival (PFS) and disease-free survival (DFS). Statistical analysis was performed to assess the association of demographic, tumor traits, and interventions with OS, PFS and DFS.</p><p><strong>Results: </strong>Only surgery followed by adjuvant therapy showed an increase in mOS [hazard ratio (HR) 0.22; 95% CI, 0.09-0.51; P<0.001), after adjustment for radiation (yes <i>vs.</i> no), resection margins (R0 <i>vs.</i> R1 or R2), and tumor location (head <i>vs.</i> body or tail). Patients who received adjuvant therapy after surgery had 2.1 times greater odds to be alive at 24 months after diagnosis than those who had surgery alone (P=0.015). PFS and DFS were not statistically significantly different among treatment groups after adjustment. Those whose disease was located in the head of the pancreas had a significantly improved OS (HR =0.27; 95% CI, 0.11-0.64; P=0.003), PFS (HR =0.40; 95% CI, 0.17-0.94; P=0.035), and DFS (HR =0.30; 95% CI, 0.13-0.67; P=0.004). Negative margins led to a significant improvement in PFS (HR =0.30; 95% CI, 0.16-0.57; P<0.001) and DFS (HR =0.30; 95% CI, 0.16-0.57; P<0.001). Those who received radiation had a non-significantly improved OS, PFS, and DFS (P>0.05).</p><p><strong>Conclusions: </strong>Our study corroborated that patients treated with adjuvant therapy after surgical resection had an mOS benefit as reported on prior phase III clinical trials. Patients with \"borderline-resectable\" pancreatic cancer are encouraged to participate in a clinical trial or clinically be treated with adjuvant therapy until more mature results from the ongoing perioperative prospective study are available.</p>","PeriodicalId":8372,"journal":{"name":"Annals of Pancreatic Cancer","volume":"3 ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2020-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7170377/pdf/nihms-1578846.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37853989","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
A. Mekapogu, S. Pothula, R. Pirola, Jeremy S. Wilson, M. Apte
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer related deaths with a dismal 5-year survival rate of only 8%. PDAC is characterised by extensive desmoplasia constituting about 50–80% of the tumour volume. Activated pancreatic stellate cells (PSC) are the major cellular source for stromal collagen; these cells drive pancreatic fibrosis and progression of PDAC. PSC are known to be activated by paracrine signals from several sources including injured epithelium, cancer cells, extracellular matrix, immune cells and nerve cells. Stromal-tumour interactions are now recognised as key processes in the development and progression of PDAC. Improved understanding of the mechanisms underlying stromal-tumour interactions may be the key for the discovery of new therapeutic targets in PDAC. This review summarises current knowledge regarding the role of PSCs in cancer biology and discusses the potential for development of novel therapeutic approaches targeting factors such as dysregulated signalling pathways, the stromal reaction itself and epigenetic changes in stromal and/or cancer cells.
{"title":"Multifunctional role of pancreatic stellate cells in pancreatic cancer","authors":"A. Mekapogu, S. Pothula, R. Pirola, Jeremy S. Wilson, M. Apte","doi":"10.21037/APC.2019.05.02","DOIUrl":"https://doi.org/10.21037/APC.2019.05.02","url":null,"abstract":"Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer related deaths with a dismal 5-year survival rate of only 8%. PDAC is characterised by extensive desmoplasia constituting about 50–80% of the tumour volume. Activated pancreatic stellate cells (PSC) are the major cellular source for stromal collagen; these cells drive pancreatic fibrosis and progression of PDAC. PSC are known to be activated by paracrine signals from several sources including injured epithelium, cancer cells, extracellular matrix, immune cells and nerve cells. Stromal-tumour interactions are now recognised as key processes in the development and progression of PDAC. Improved understanding of the mechanisms underlying stromal-tumour interactions may be the key for the discovery of new therapeutic targets in PDAC. This review summarises current knowledge regarding the role of PSCs in cancer biology and discusses the potential for development of novel therapeutic approaches targeting factors such as dysregulated signalling pathways, the stromal reaction itself and epigenetic changes in stromal and/or cancer cells.","PeriodicalId":8372,"journal":{"name":"Annals of Pancreatic Cancer","volume":"197 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"74506921","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Immunotherapy can take advantage of the immunogenic response that chemotherapy elicits in tumors. Gemcitabine is a standard agent used in the treatment of pancreatic cancer, with known effects on the tumor immune microenvironment. The combination immunotherapy of the GVAX cancer vaccine, anti-PD-1 antibody and anti-CSF-1R antibody has been shown to improve survival in a murine model of metastatic pancreatic adenocarcinoma. This combination regimen also increased the infiltration of CD8+ T-cells that expressed both PD1 and CD137, and these T-cells were shown to express high levels of interferon-gamma, a marker of cytotoxic effector CD8+ T-cells. The effect of the addition of gemcitabine to this promising immunotherapy regimen has not been investigated.
Methods: Mice with liver-metastatic pancreatic adenocarcinoma were followed for 120 days to determine if adding immunotherapy, which comprised of varying combinations of GVAX, anti-PD-1 antibody and anti-CSF-1R antibody, to gemcitabine improved survival. Tumor-infiltrating CD8+ T-cells and myeloid cells, harvested after the mice were treated for 2 weeks, were analyzed with flow cytometry to characterize the effect the chemo-immunotherapy regimen had on the tumor microenvironment (TME).
Results: Adding combination immunotherapy after gemcitabine improved survival compared to gemcitabine treatment alone (gemcitabine/GVAX/anti-PD1, P<0.001; gemcitabine/anti-PD1/anti-CSF-1R, P<0.05; gemcitabine/GVAX/anti-PD1/anti-CSF-1R, P<0.01). However, there was no difference in survival between the three chemo-immunotherapy treatment regimens. Compared to gemcitabine-only treatment, the chemo-immunotherapy regimens also increased the percentage of tumor-infiltrating CD8+ T-cells that expressed interferon-gamma (gemcitabine/GVAX/anti-PD1, P<0.0001 and gemcitabine/GVAX/anti-PD1/anti-CSF-1R, P<0.0001). The chemo-immunotherapy regimens also increased the number of tumor-infiltrating PD1+CD137+CD8+ T-cells and interferon-gamma-expressing PD1+CD137+CD8+ T-cells, but these increases were not statistically significant. Anti-CSF-1R antibody decreased the infiltration of myeloid cells and myeloid-derived suppressor cells caused by GVAX (P<0.05), and trended towards decreasing tumor-associated macrophages (TAMs) (P=0.18).
Conclusions: The addition of anti-PD1 antibody with GVAX and/or anti-CSF-1R antibody to gemcitabine improved the survival of mice with liver-metastatic pancreatic ductal adenocarcinoma (PDA). Gemcitabine with GVAX and anti-PD1 with or without anti-CSF-1R also improved the infiltration of effector CD8+ T-cells, and the presence of anti-CSF-1R in the chemo-immunotherapy regimens decreased the infiltration of myeloid cells. The overlapping mechanisms of the components in the chemo-immunotherapy regimens may explain the lack of survival difference between the various regimens, and this remains to be explored.
背景:免疫疗法可以利用化疗在肿瘤中引起的免疫原性反应。吉西他滨是治疗胰腺癌的标准药物,已知会对肿瘤免疫微环境产生影响。GVAX 癌症疫苗、抗-PD-1 抗体和抗-CSF-1R 抗体的联合免疫疗法在转移性胰腺腺癌小鼠模型中改善了生存率。这种联合疗法还增加了同时表达 PD1 和 CD137 的 CD8+ T 细胞的浸润,这些 T 细胞被证明表达高水平的干扰素-γ,这是细胞毒性效应 CD8+ T 细胞的标志。在这一前景看好的免疫疗法方案中加入吉西他滨的效果尚未得到研究:方法:对肝转移性胰腺腺癌小鼠进行为期120天的随访,以确定在吉西他滨的基础上添加免疫疗法(包括GVAX、抗PD-1抗体和抗CSF-1R抗体的不同组合)是否能提高生存率。小鼠接受治疗2周后收获的肿瘤浸润CD8+ T细胞和髓系细胞通过流式细胞术进行了分析,以确定化疗免疫疗法对肿瘤微环境(TME)的影响:结果:与单用吉西他滨治疗相比,在吉西他滨治疗后加用联合免疫疗法可提高生存率(吉西他滨/GVAX/抗-PD1,PConclusions:在吉西他滨基础上添加抗PD1抗体和GVAX和/或抗CSF-1R抗体可提高肝转移性胰腺导管腺癌(PDA)小鼠的生存率。吉西他滨与 GVAX 和抗-PD1(含或不含抗-CSF-1R)也改善了效应 CD8+ T 细胞的浸润,而化疗免疫疗法中抗-CSF-1R 的存在减少了骨髓细胞的浸润。化疗-免疫治疗方案中各成分的重叠机制可能是各种方案之间缺乏生存差异的原因,这一点仍有待探讨。
{"title":"Adding combination immunotherapy consisting of cancer vaccine, anti-PD-1 and anti-CSF1R antibodies to gemcitabine improves anti-tumor efficacy in murine model of pancreatic ductal adenocarcinoma.","authors":"May Tun Saung, Lei Zheng","doi":"10.21037/apc.2019.11.01","DOIUrl":"10.21037/apc.2019.11.01","url":null,"abstract":"<p><strong>Background: </strong>Immunotherapy can take advantage of the immunogenic response that chemotherapy elicits in tumors. Gemcitabine is a standard agent used in the treatment of pancreatic cancer, with known effects on the tumor immune microenvironment. The combination immunotherapy of the GVAX cancer vaccine, anti-PD-1 antibody and anti-CSF-1R antibody has been shown to improve survival in a murine model of metastatic pancreatic adenocarcinoma. This combination regimen also increased the infiltration of CD8+ T-cells that expressed both PD1 and CD137, and these T-cells were shown to express high levels of interferon-gamma, a marker of cytotoxic effector CD8+ T-cells. The effect of the addition of gemcitabine to this promising immunotherapy regimen has not been investigated.</p><p><strong>Methods: </strong>Mice with liver-metastatic pancreatic adenocarcinoma were followed for 120 days to determine if adding immunotherapy, which comprised of varying combinations of GVAX, anti-PD-1 antibody and anti-CSF-1R antibody, to gemcitabine improved survival. Tumor-infiltrating CD8+ T-cells and myeloid cells, harvested after the mice were treated for 2 weeks, were analyzed with flow cytometry to characterize the effect the chemo-immunotherapy regimen had on the tumor microenvironment (TME).</p><p><strong>Results: </strong>Adding combination immunotherapy after gemcitabine improved survival compared to gemcitabine treatment alone (gemcitabine/GVAX/anti-PD1, P<0.001; gemcitabine/anti-PD1/anti-CSF-1R, P<0.05; gemcitabine/GVAX/anti-PD1/anti-CSF-1R, P<0.01). However, there was no difference in survival between the three chemo-immunotherapy treatment regimens. Compared to gemcitabine-only treatment, the chemo-immunotherapy regimens also increased the percentage of tumor-infiltrating CD8+ T-cells that expressed interferon-gamma (gemcitabine/GVAX/anti-PD1, P<0.0001 and gemcitabine/GVAX/anti-PD1/anti-CSF-1R, P<0.0001). The chemo-immunotherapy regimens also increased the number of tumor-infiltrating PD1+CD137+CD8+ T-cells and interferon-gamma-expressing PD1+CD137+CD8+ T-cells, but these increases were not statistically significant. Anti-CSF-1R antibody decreased the infiltration of myeloid cells and myeloid-derived suppressor cells caused by GVAX (P<0.05), and trended towards decreasing tumor-associated macrophages (TAMs) (P=0.18).</p><p><strong>Conclusions: </strong>The addition of anti-PD1 antibody with GVAX and/or anti-CSF-1R antibody to gemcitabine improved the survival of mice with liver-metastatic pancreatic ductal adenocarcinoma (PDA). Gemcitabine with GVAX and anti-PD1 with or without anti-CSF-1R also improved the infiltration of effector CD8+ T-cells, and the presence of anti-CSF-1R in the chemo-immunotherapy regimens decreased the infiltration of myeloid cells. The overlapping mechanisms of the components in the chemo-immunotherapy regimens may explain the lack of survival difference between the various regimens, and this remains to be explored.</p>","PeriodicalId":8372,"journal":{"name":"Annals of Pancreatic Cancer","volume":"2 ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7220030/pdf/nihms-1586273.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37932770","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Alejandro Recio-Boiles, Aparna Nallagangula, Summana Veeravelli, Jessica Vondrak, K. Saboda, D. Roe, E. Elquza, A. McBride, H. Babiker
Background Post-surgical pathology (SP) staging correlates with long-term survival. Neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) have been shown to predict prognosis and extent of tumor in patients with metastatic pancreatic ductal adenocarcinoma (PDAC). This study aimed to correlate NLR and PLR to radiological clinical staging (CS), carbohydrate antigen (CA) 19-9 tumor marker and SP staging in patients with resectable-PDAC (R-PDAC); and to investigate NLR and PLR as potential markers to guide neoadjuvant therapy. Methods Data were collected retrospectively from R-PDAC patients who received upfront surgery from November 2011 to December 2016. NLR and PLR values on the day of diagnosis and surgery were collected. SP, tumor size, location, resected margins (RM), lymphovascular/perineural invasion (LVI/PNI), lymph node involvement, and AJCC/TNM 8th Edition staging were obtained. Associations were assessed using linear, ordinal logistic, and poison regressions or Kruskal Willis Rank Sum Test per the nature of outcome variables, with statistical significance at p-value <0.05. Results Fifty-five patients were identified with resectable stage I (61%) and II (38%). They had a mean age of 66 years (48-87 years) and were 47.2% male, 83.6% white, 90.9% non-Hispanic and 89% with ECOG 0-1. NLR/PLR at diagnosis for R0, R1 and R2 were 6.7/241, 4.8/224, and 2.9/147 (P=0.01/0.002), respectively. NLR/PLR for N0 and N1 were 5.1/212 and 2.7/138.3 (P=0.03/0.009) at diagnosis. No other significant association was detected. Conclusions These findings suggest that NLR/PLR inversely correlates with RM and lymph node status in patients with R-PDAC, but require prospective evaluation in clinically defined scenarios.
{"title":"Neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios inversely correlate to clinical and pathologic stage in patients with resectable pancreatic ductal adenocarcinoma.","authors":"Alejandro Recio-Boiles, Aparna Nallagangula, Summana Veeravelli, Jessica Vondrak, K. Saboda, D. Roe, E. Elquza, A. McBride, H. Babiker","doi":"10.21037/APC.2019.06.01","DOIUrl":"https://doi.org/10.21037/APC.2019.06.01","url":null,"abstract":"Background\u0000Post-surgical pathology (SP) staging correlates with long-term survival. Neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) have been shown to predict prognosis and extent of tumor in patients with metastatic pancreatic ductal adenocarcinoma (PDAC). This study aimed to correlate NLR and PLR to radiological clinical staging (CS), carbohydrate antigen (CA) 19-9 tumor marker and SP staging in patients with resectable-PDAC (R-PDAC); and to investigate NLR and PLR as potential markers to guide neoadjuvant therapy.\u0000\u0000\u0000Methods\u0000Data were collected retrospectively from R-PDAC patients who received upfront surgery from November 2011 to December 2016. NLR and PLR values on the day of diagnosis and surgery were collected. SP, tumor size, location, resected margins (RM), lymphovascular/perineural invasion (LVI/PNI), lymph node involvement, and AJCC/TNM 8th Edition staging were obtained. Associations were assessed using linear, ordinal logistic, and poison regressions or Kruskal Willis Rank Sum Test per the nature of outcome variables, with statistical significance at p-value <0.05.\u0000\u0000\u0000Results\u0000Fifty-five patients were identified with resectable stage I (61%) and II (38%). They had a mean age of 66 years (48-87 years) and were 47.2% male, 83.6% white, 90.9% non-Hispanic and 89% with ECOG 0-1. NLR/PLR at diagnosis for R0, R1 and R2 were 6.7/241, 4.8/224, and 2.9/147 (P=0.01/0.002), respectively. NLR/PLR for N0 and N1 were 5.1/212 and 2.7/138.3 (P=0.03/0.009) at diagnosis. No other significant association was detected.\u0000\u0000\u0000Conclusions\u0000These findings suggest that NLR/PLR inversely correlates with RM and lymph node status in patients with R-PDAC, but require prospective evaluation in clinically defined scenarios.","PeriodicalId":8372,"journal":{"name":"Annals of Pancreatic Cancer","volume":"88 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"86790753","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Benjamin O. Lawson, R. Hultsch, L. Caldwell, K. Gosselin, G. Jameson, E. Borazanci
Background: Thrombocytopenia may be a concern in treating pancreatic ductal adenocarcinomas (PDAC). Due to anatomic position these tumors commonly cause narrowing or occlusion of the splenic vein which may lead to hypersplenism and thrombocytopenia due to sequestration. PDAC patients with thrombocytopenia have limited options for treatment. Partial splenic embolization (PSE) is a procedure developed as an alternative to splenectomy in individuals with hypersplenism. The purpose of our study was to review outcomes of PSE on thrombocytopenia in stage III and IV PDAC patients. Methods: From November 2015 through January 2018, we conducted a retrospective chart analysis of 8 patients with stage III or stage IV pancreatic cancer who had undergone PSE. The primary objective of this retrospective study was to understand the utility of PSE in treating thrombocytopenia in pancreatic cancer patients by the effect on the individual’s platelet count and the subsequent exposure to chemotherapy treatment. Specific demographic data points were recorded including date of diagnosis, stage, location of primary origin, and site of metastasis. Other data including hospital days post-embolization, post-embolization syndrome (PES), days to return of chemotherapy, survival, and pre/post platelet counts at designated intervals were reviewed. Results: Seven-eighths patients were diagnosed with stage IV pancreatic adenocarcinoma with the pancreatic head being the most common primary site (50%). Most common site of metastasis was liver. PES was found in 5/8 patients with the average number of hospital days after the procedure 1.38 (SD =1.06). Mean platelet count pre-splenic embolization was 93.00 (SD =12.59). One-week post-embolization mean platelet count was 147.00 (SD =69.60). Four-week ( χ =183.60, SD =64.93), six-week ( χ =148.40, SD =40.58), and three-month ( χ =161.00, SD =79.07) intervals were used to further assess platelet change. The results of the ANOVA were significant, F (4) =3.65, P=0.027, =0.48. Post-hoc analyses revealed significant differences between one-week and four-week post-embolization (P=0.008). Time to restarting chemotherapy ranged from 1 to 129 days with an average day to restarting chemotherapy of 24.12 (SD =42.70). The median overall survival was 7.22 months. Conclusions: In considering our study’s small sample size, PSE should be considered a safe approach in managing thrombocytopenia long-term in stage III or stage IV PDAC patients. PSE may allow for further chemotherapy to improve overall survival.
{"title":"Partial splenic embolization to alleviate thrombocytopenia in stage III and IV pancreatic ductal adenocarcinoma patients","authors":"Benjamin O. Lawson, R. Hultsch, L. Caldwell, K. Gosselin, G. Jameson, E. Borazanci","doi":"10.21037/APC.2019.05.01","DOIUrl":"https://doi.org/10.21037/APC.2019.05.01","url":null,"abstract":"Background: Thrombocytopenia may be a concern in treating pancreatic ductal adenocarcinomas (PDAC). Due to anatomic position these tumors commonly cause narrowing or occlusion of the splenic vein which may lead to hypersplenism and thrombocytopenia due to sequestration. PDAC patients with thrombocytopenia have limited options for treatment. Partial splenic embolization (PSE) is a procedure developed as an alternative to splenectomy in individuals with hypersplenism. The purpose of our study was to review outcomes of PSE on thrombocytopenia in stage III and IV PDAC patients. \u0000 Methods: From November 2015 through January 2018, we conducted a retrospective chart analysis of 8 patients with stage III or stage IV pancreatic cancer who had undergone PSE. The primary objective of this retrospective study was to understand the utility of PSE in treating thrombocytopenia in pancreatic cancer patients by the effect on the individual’s platelet count and the subsequent exposure to chemotherapy treatment. Specific demographic data points were recorded including date of diagnosis, stage, location of primary origin, and site of metastasis. Other data including hospital days post-embolization, post-embolization syndrome (PES), days to return of chemotherapy, survival, and pre/post platelet counts at designated intervals were reviewed. \u0000 Results: Seven-eighths patients were diagnosed with stage IV pancreatic adenocarcinoma with the pancreatic head being the most common primary site (50%). Most common site of metastasis was liver. PES was found in 5/8 patients with the average number of hospital days after the procedure 1.38 (SD =1.06). Mean platelet count pre-splenic embolization was 93.00 (SD =12.59). One-week post-embolization mean platelet count was 147.00 (SD =69.60). Four-week ( χ =183.60, SD =64.93), six-week ( χ =148.40, SD =40.58), and three-month ( χ =161.00, SD =79.07) intervals were used to further assess platelet change. The results of the ANOVA were significant, F (4) =3.65, P=0.027, =0.48. Post-hoc analyses revealed significant differences between one-week and four-week post-embolization (P=0.008). Time to restarting chemotherapy ranged from 1 to 129 days with an average day to restarting chemotherapy of 24.12 (SD =42.70). The median overall survival was 7.22 months. \u0000 Conclusions: In considering our study’s small sample size, PSE should be considered a safe approach in managing thrombocytopenia long-term in stage III or stage IV PDAC patients. PSE may allow for further chemotherapy to improve overall survival.","PeriodicalId":8372,"journal":{"name":"Annals of Pancreatic Cancer","volume":"34 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"88699681","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Application of the CARE guideline as reporting standard in the Annals of Pancreatic Cancer","authors":"","doi":"10.21037/apc.2019.10.02","DOIUrl":"https://doi.org/10.21037/apc.2019.10.02","url":null,"abstract":"","PeriodicalId":8372,"journal":{"name":"Annals of Pancreatic Cancer","volume":"19 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"82119798","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
In 2018, there was an estimated 55,440 new cases of pancreatic adenocarcinoma (PDAC) in the United States. Generally, most new cases are in an advanced stage, usually due to the lack of symptoms during the early stages of the disease. Currently, the vast majority of therapeutic regimens have shown only modest effects in this setting, and median survival ranges from 6 to 11 months. Indeed, better therapies for these patients are urgently needed. Epigenetics refers to the somatically heritable differences in gene expression not attributable to intrinsic alterations in the primary sequence of DNA. Core elements of the epigenetic regulation of gene expression include how DNA is packaged around nucleosomes, how chromatin and nucleosomes are modified by a complex series of enzymes and their subsequent interactions with proteins that recognize these modifications. The recognition of the essential role of epigenetic alterations in the development and progression of PDAC has revolutionized our knowledge of this disease and has immediate translational implications for targeting epigenetic abnormalities in PDAC for therapeutic purposes. Moreover, recent work with epigenetic modulatory drugs (EMDs) has shown that these agents may be capable of altering the immunogenicity of the tumor microenvironment (TME), to reverse immune suppression and to ‘prime’ tumors for immunotherapy. This review summarizes the current knowledge of epigenetic alterations in PDAC with a focus on the translational application of targeting epigenetic-based events as new therapeutic approach for this disease.
{"title":"Targeting the epigenome of pancreatic cancer for therapy: challenges and opportunities","authors":"M. Baretti, N. Ahuja, N. Azad","doi":"10.21037/apc.2019.10.01","DOIUrl":"https://doi.org/10.21037/apc.2019.10.01","url":null,"abstract":"In 2018, there was an estimated 55,440 new cases of pancreatic adenocarcinoma (PDAC) in the United States. Generally, most new cases are in an advanced stage, usually due to the lack of symptoms during the early stages of the disease. Currently, the vast majority of therapeutic regimens have shown only modest effects in this setting, and median survival ranges from 6 to 11 months. Indeed, better therapies for these patients are urgently needed. Epigenetics refers to the somatically heritable differences in gene expression not attributable to intrinsic alterations in the primary sequence of DNA. Core elements of the epigenetic regulation of gene expression include how DNA is packaged around nucleosomes, how chromatin and nucleosomes are modified by a complex series of enzymes and their subsequent interactions with proteins that recognize these modifications. The recognition of the essential role of epigenetic alterations in the development and progression of PDAC has revolutionized our knowledge of this disease and has immediate translational implications for targeting epigenetic abnormalities in PDAC for therapeutic purposes. Moreover, recent work with epigenetic modulatory drugs (EMDs) has shown that these agents may be capable of altering the immunogenicity of the tumor microenvironment (TME), to reverse immune suppression and to ‘prime’ tumors for immunotherapy. This review summarizes the current knowledge of epigenetic alterations in PDAC with a focus on the translational application of targeting epigenetic-based events as new therapeutic approach for this disease.","PeriodicalId":8372,"journal":{"name":"Annals of Pancreatic Cancer","volume":"64 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"77231898","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-10-01Epub Date: 2019-10-23DOI: 10.21037/apc.2019.09.01
Imani Bijou, Jin Wang
Despite advances in translational research, the overall 5-year survival for pancreatic cancer remains dismal and with rising incidence pancreatic cancer is predicted to be the second leading cause of cancer death for many developed countries. Surgical intervention followed by cytotoxic chemotherapy are currently the best options for treatment, but disease recurrence is very common. Efforts to develop new therapeutic agents and delivery systems are necessary to achieve better clinical efficacy with less toxicity. Promising prospects are arising with new preclinical and clinical therapeutic strategies using small molecule targeted therapies, RNAi, stromal therapies, and immunotherapies. With a better understanding of the biology to aid target selection and discovery of biomarkers to aid precision medicine, better opportunities will evolve to shape the therapeutic landscape, enhance patient quality of life and increase overall survival.
{"title":"Evolving trends in pancreatic cancer therapeutic development.","authors":"Imani Bijou, Jin Wang","doi":"10.21037/apc.2019.09.01","DOIUrl":"10.21037/apc.2019.09.01","url":null,"abstract":"<p><p>Despite advances in translational research, the overall 5-year survival for pancreatic cancer remains dismal and with rising incidence pancreatic cancer is predicted to be the second leading cause of cancer death for many developed countries. Surgical intervention followed by cytotoxic chemotherapy are currently the best options for treatment, but disease recurrence is very common. Efforts to develop new therapeutic agents and delivery systems are necessary to achieve better clinical efficacy with less toxicity. Promising prospects are arising with new preclinical and clinical therapeutic strategies using small molecule targeted therapies, RNAi, stromal therapies, and immunotherapies. With a better understanding of the biology to aid target selection and discovery of biomarkers to aid precision medicine, better opportunities will evolve to shape the therapeutic landscape, enhance patient quality of life and increase overall survival.</p>","PeriodicalId":8372,"journal":{"name":"Annals of Pancreatic Cancer","volume":"2 ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7575122/pdf/nihms-1069278.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"38519395","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}