1
|
Cui J, Qin S, Zhou Y, Zhang S, Sun X, Zhang M, Cui J, Fang W, Gu K, Li Z, Wang J, Chen X, Yao J, Zhou J, Wang G, Bai Y, Xiao J, Qiu W, Wang B, Xia T, Wang C, Kong L, Yin J, Zhang T, Shen X, Fu D, Gao C, Wang H, Wang Q, Wang L. Irinotecan hydrochloride liposome HR070803 in combination with 5-fluorouracil and leucovorin in locally advanced or metastatic pancreatic ductal adenocarcinoma following prior gemcitabine-based therapy (PAN-HEROIC-1): a phase 3 trial. Signal Transduct Target Ther 2024; 9:248. [PMID: 39300077 DOI: 10.1038/s41392-024-01948-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/15/2024] [Accepted: 08/14/2024] [Indexed: 09/22/2024] Open
Abstract
Liposomal irinotecan has shown promising antitumor activity in patients with advanced or metastatic pancreatic ductal adenocarcinoma (PDAC) who have undergone prior gemcitabine-based therapies. This randomized, double-blind, parallel-controlled, multicenter phase 3 study (NCT05074589) assessed the efficacy and safety of liposomal irinotecan HR070803 combined with 5-fluorouracil (5-FU) and leucovorin (LV) in this patient population. Patients with unresectable, locally advanced, or metastatic PDAC who had previously received gemcitabine-based therapies were randomized 1:1 to receive either HR070803 (60 mg/m2 anhydrous irinotecan hydrochloride, equal to 56.5 mg/m2 free base) or placebo, both in combination with 5-FU (2000 mg/m2) and LV (200 mg/m2), all given intravenously every two weeks. The primary endpoint of the study was overall survival (OS). A total of 298 patients were enrolled and received HR070803 plus 5-FU/LV (HR070803 group, n = 149) or placebo plus 5-FU/LV (placebo group, n = 149). Median OS was significantly improved in the HR070803 group compared to the placebo group (7.4 months [95% CI 6.1-8.4] versus 5.0 months [95% CI 4.3-6.0]; HR 0.63 [95% CI 0.48-0.84]; two-sided p = 0.0019). The most common grade ≥ 3 adverse events in the HR070803 group were increased gamma-glutamyltransferase (19.0% versus 11.6% in placebo group) and decreased neutrophil count (12.9% versus 0 in placebo group). No treatment-related deaths occurred in the HR070803 group, while the placebo group reported one treatment-related death (abdominal infection). HR070803 in combination with 5-FU/LV has shown promising efficacy and manageable safety in advanced or metastatic PDAC in the second-line setting, representing a potential option in this patient population.
Collapse
Affiliation(s)
- Jiujie Cui
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shukui Qin
- GI Cancer Center, Nanjing Tianyinshan Hospital, China Pharmaceutical University, Nanjing, China.
| | - Yuhong Zhou
- Medical Oncology, Zhongshan Hospital Affiliated with Fudan University, Shanghai, China
| | - Shuang Zhang
- Department of Biotherapy, West China School of Medicine/West China Hospital of Sichuan University, Chengdu, China
| | - Xiaofeng Sun
- Internal Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Mingjun Zhang
- Medical Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiuwei Cui
- Department of Oncology, The First Hospital of Jilin University, Changchun, China
| | - Weijia Fang
- Medical Oncology III, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kangsheng Gu
- Medical Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhihua Li
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jufeng Wang
- Department of Oncology, The Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, China
| | - Xiaobing Chen
- Department of Oncology, The Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, China
| | - Jun Yao
- Department of Oncology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Jun Zhou
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Gang Wang
- Department of Oncology, The First Affiliated Hospital of USTC Anhui Provincial Hospital, Hefei, China
| | - Yuxian Bai
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Juxiang Xiao
- Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wensheng Qiu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bangmao Wang
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Xia
- Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Chunyue Wang
- Medical Oncology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Li Kong
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jiajun Yin
- General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xionghu Shen
- Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Deliang Fu
- Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuntao Gao
- Pancreatic Surgery, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Huan Wang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Quanren Wang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Liwei Wang
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
2
|
Li Y, Xu C, Han H, Pascual-Sabater S, Fillat C, Goel A. Aronia Berry Extract Modulates MYD88/NF-kB/P-Glycoprotein Axis to Overcome Gemcitabine Resistance in Pancreatic Cancer. Pharmaceuticals (Basel) 2024; 17:911. [PMID: 39065761 PMCID: PMC11279572 DOI: 10.3390/ph17070911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease with poor survival rates, primarily due to the limited effectiveness of gemcitabine (Gem)-based chemotherapy, as well as the acquisition of chemotherapeutic resistance. Aronia berry extracts (ABEs), abundant in phenolic constituents, have been recently recognized for their anticancer properties as well as their encouraging potential to help overcome chemoresistance in various cancers. In the present study, we explored ABE's potential to overcome Gem resistance in PDAC and identify specific growth regulatory pathways responsible for its anticancer activity. Through a series of in vitro experiments in gemcitabine-resistant (Gem-R) cells, we elucidated the synergistic interactions between Gem and ABE treatments. Using advanced transcriptomic analysis and network pharmacology, we revealed key molecular pathways linked to chemoresistance and potential therapeutic targets of ABE in Gem-R PDAC cells. Subsequently, the findings from cell culture studies were validated in patient-derived 3D tumor organoids (PDOs). The combination treatment of ABE and Gem demonstrated significant synergism and anticancer effects on cell viability, proliferation, migration, and invasion in Gem-R cells. Transcriptomic analysis revealed a correlation between the NF-Κb signaling pathway and Gem-R (p < 0.05), exhibiting a marked upregulation of MYD88. Additionally, MYD88 exhibited a significant correlation with the overall survival rates in patients with PDAC patients in the TCGA cohort (HR = 1.58, p < 0.05). The MYD88/NF-Κb pathway contributes to chemoresistance by potentially upregulating efflux transporters like P-glycoprotein (P-gp). Our findings revealed that the combined treatment with ABE suppressed the NF-Κb pathway by targeting MYD88 and reducing P-gp expression to overcome Gem resistance. Lastly, the combination therapy proved highly effective in PDOs in reducing both their number and size (p < 0.05). Our study offers previously unrecognized insights into the ability of ABE to overcome Gem resistance in PDAC cells through its targeting of the MYD88/NF-κb/P-gp axis, hence providing a safe and cost-effective adjunctive therapeutic strategy to improve treatment outcomes in PDAC.
Collapse
Affiliation(s)
- Yuan Li
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA 91016, USA; (Y.L.); (C.X.)
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, China
| | - Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA 91016, USA; (Y.L.); (C.X.)
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116004, China
| | - Haiyong Han
- Division of Molecular Medicine, The Translational Genomics Research Institute, Phoenix, AZ 85004, USA;
| | - Silvia Pascual-Sabater
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-S.); (C.F.)
| | - Cristina Fillat
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-S.); (C.F.)
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA 91016, USA; (Y.L.); (C.X.)
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
3
|
Yu S, Wang M, Zhang H, Guo X, Qin R. Resistance to gemcitabine is mediated by the circ_0036627/miR-145/S100A16 axis in pancreatic cancer. J Cell Mol Med 2024; 28:e18444. [PMID: 38924205 PMCID: PMC11196374 DOI: 10.1111/jcmm.18444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 04/27/2024] [Accepted: 05/11/2024] [Indexed: 06/28/2024] Open
Abstract
The development of gemcitabine (GEM) resistance severely limits the treatment efficacy in pancreatic cancer (PC) and increasing evidence highlights the vital roles of circular RNAs (circRNAs) in the tumorigenesis, progression and drug resistance of PC. However, the circRNAs underlying GEM resistance development of PC remains to be clarified. The current research aims to unveil the roles of circ_0036627 in dictating the aggressiveness and GEM sensitivity in PC. We reported the increased expression of circ_0036627 in PC tissues and PC cell lines. Elevated circ_0036627 expression level was correlated with advanced tumour grade and poor overall survival in PC patients. Functional assays and in vivo experiments demonstrated that circ_0036627 overexpression was required for the proliferation, migration invasion and GEM resistance in PC cells. circ_0036627 knockdown suppressed tumour development in vivo. The molecular analysis further showed that circ_0036627 increased S100A16 expression by sponging microRNA-145 (miR-145), a tumour-suppressive miRNA that could significantly attenuate PC cell proliferation, migration, invasion and GEM resistance. Furthermore, our findings suggested that S100A16 acted as an oncogenic factor to promote aggressiveness and GEM resistance in PC cells. In conclusion, the current findings provide new mechanistic insights into PC aggressiveness and GEM resistance, suggesting the critical role of circ_0036627/miR-145/S100A16 axis in PC progression and drug resistance development and offering novel therapeutic targets for PC therapy.
Collapse
Affiliation(s)
- Shuo Yu
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Min Wang
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Hang Zhang
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Xingjun Guo
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Renyi Qin
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
4
|
Long SA, Amparo AM, Goodhart G, Ahmad SA, Waters AM. Evaluation of KRAS inhibitor-directed therapies for pancreatic cancer treatment. Front Oncol 2024; 14:1402128. [PMID: 38800401 PMCID: PMC11116577 DOI: 10.3389/fonc.2024.1402128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Despite significant advancements in the treatment of other cancers, pancreatic ductal adenocarcinoma (PDAC) remains one of the world's deadliest cancers. More than 90% of PDAC patients harbor a Kirsten rat sarcoma (KRAS) gene mutation. Although the clinical potential of anti-KRAS therapies has long been realized, all initial efforts to target KRAS were unsuccessful. However, with the recent development of a new generation of KRAS-targeting drugs, multiple KRAS-targeted treatment options for patients with PDAC have entered clinical trials. In this review, we provide an overview of current standard of care treatment, describe RAS signaling and the relevance of KRAS mutations, and discuss RAS isoform- and mutation-specific differences. We also evaluate the clinical efficacy and safety of mutation-selective and multi-selective inhibitors, in the context of PDAC. We then provide a comparison of clinically relevant KRAS inhibitors to second-line PDAC treatment options. Finally, we discuss putative resistance mechanisms that may limit the clinical effectiveness of KRAS-targeted therapies and provide a brief overview of promising therapeutic approaches in development that are focused on mitigating these resistance mechanisms.
Collapse
Affiliation(s)
- Szu-Aun Long
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Amber M. Amparo
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Grace Goodhart
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Syed A. Ahmad
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Andrew M. Waters
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
5
|
Pourali G, Donyadideh G, Mehrabadi S, Hamid F, Hassanian SM, Ferns GA, Khazaei M, Avan A. Clinical practice guidelines for interventional treatment of pancreatic cancer. RECENT ADVANCES IN NANOCARRIERS FOR PANCREATIC CANCER THERAPY 2024:345-373. [DOI: 10.1016/b978-0-443-19142-8.00008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
6
|
Amin K, Khan H, Hearld LR, Chu DI, Prete V, Mehari KR, Heslin MJ, Fonseca AL. Association between Rural Residence and Processes of Care in Pancreatic Cancer Treatment. J Gastrointest Surg 2023; 27:2155-2165. [PMID: 37553515 PMCID: PMC10731615 DOI: 10.1007/s11605-023-05764-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/17/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND Pancreatic adenocarcinoma (PDAC) is an aggressive malignancy associated with poor outcomes. Surgical resection and receipt of multimodal therapy have been shown to improve outcomes in patients with potentially resectable PDAC; however treatment and outcome disparities persist on many fronts. The aim of this study was to analyze the relationship between rural residence and receipt of quality cancer care in patients diagnosed with non-metastatic PDAC. METHODS Using the National Cancer Database, patients with non-metastatic pancreatic cancer were identified from 2006-2016. Patients were classified as living in metropolitan, urban, or rural areas. Multivariable logistic regression was used to identify predictors of cancer treatment and survival. RESULTS A total of 41,786 patients were identified: 81.6% metropolitan, 16.2% urban, and 2.2% rural. Rural residing patients were less likely to receive curative-intent surgery (p = 0.037) and multimodal therapy (p < 0.001) compared to their metropolitan and urban counterparts. On logistic regression analysis, rural residence was independently associated with decreased surgical resection [OR 0.82; CI 95% 0.69-0.99; p = 0.039] and multimodal therapy [OR 0.70; CI 95% 0.38-0.97; p = 0.047]. Rural residence independently predicted decreased overall survival [OR 1.64; CI 95% 1.45-1.93; p < 0.001] for all patients that were analyzed. In the cohort of patients who underwent surgical resection, rural residence did not independently predict overall survival [OR 0.97; CI 95% 0.85-1.11; p = 0.652]. CONCLUSIONS Rural residence impacts receipt of optimal cancer care in patients with non-metastatic PDAC but does not predict overall survival in patients who receive curative-intent treatment.
Collapse
Affiliation(s)
- Krisha Amin
- Department of Surgery, The University of South Alabama, 2451 USA Medical Center Drive, Mastin, 705, Mobile, AL, 36617, USA
| | - Hamza Khan
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Larry R Hearld
- Department of Health Services Administration, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Daniel I Chu
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victoria Prete
- Department of Surgery, The University of South Alabama, 2451 USA Medical Center Drive, Mastin, 705, Mobile, AL, 36617, USA
| | - Krista R Mehari
- Department of Psychology, The University of South Alabama, Mobile, AL, USA
| | - Martin J Heslin
- Department of Surgery, The University of South Alabama, 2451 USA Medical Center Drive, Mastin, 705, Mobile, AL, 36617, USA
| | - Annabelle L Fonseca
- Department of Surgery, The University of South Alabama, 2451 USA Medical Center Drive, Mastin, 705, Mobile, AL, 36617, USA.
| |
Collapse
|
7
|
Varzaru B, Iacob RA, Croitoru AE, Iacob SM, Radu CE, Dumitrescu SM, Gheorghe C. Real-Life Results of Palliative Chemotherapy in Metastatic Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:3500. [PMID: 37444612 DOI: 10.3390/cancers15133500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
PURPOSE To assess the efficacy of FOLFIRINOX(FFX), gemcitabine-based regimens (GB), and gemcitabine monotherapy (Gem) in patients with metastatic pancreatic ductal adenocarcinoma (mPDAC). METHODS This is a retrospective study that included 83 patients with mPDAC treated with first-line chemotherapy (L1) with either FFX, GB or Gem between 2015 and 2017. Progression-free survival (PFS) for L1 and second-line chemotherapy (L2) (PFS-L1 and PFS-L2) and overall survival (OS) were estimated using the Kaplan-Meier method. RESULTS Median PFS-L1 for FFX, GB and Gem groups was 9 months (95% (Confidence Interval) CI 2.76-15.24), 5 months (95%CI 3.44-6.56), and 5 months (95%CI 3.76-6.24), respectively (p = 0.04). OS was 14 months (95%CI 11.16-16.85), 12 months (95%CI: 9.44-11.56), and 7 months (95%CI: 5.7-8.3) for patients treated with FFX, GB, and Gem, respectively (p = 0.0001). ECOG-PS (0/1) (Hazard Ratio (HR) 6.74, p = 0.002), age > 70 years (HR 0.25, p = 0.04), body tumors (HR 2.8, p = 0.048), CA19-9 > 39 U/mL (HR 0.26, p = 0.02), and neutrophil-to-lymphocyte ratio (NLR) > 4.15 (HR 6.76, p = 0.001) were independent prognostic factors for PFS-L1. Male gender (HR 3.02, p = 0.026), ECOG-PS (0/1) (HR 4.21, p = 0.003), L1 with FFX (HR 0.255, p = 0.007), and NLR > 4.15 (HR 2.65, p = 0.04) were independent prognostic factors of OS. PFS-L2 (HR 6.91, p = 0.013) and OS-L2 (HR 6.95, p = 0.037) were significantly higher in patients first treated with FFX. CONCLUSIONS The OS of patients who receive FFX or GB is comparable. The best PFS-L1 belongs to the FFX group. Male gender, ECOG-PS 0/1, the FFX regimen, and NLR > 4.15 were independent predictors of OS. PFS-L2 and OS-L2 were favorably impacted by L1 with FFX.
Collapse
Affiliation(s)
- Bianca Varzaru
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| | - Razvan A Iacob
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, 022238 Bucharest, Romania
| | - Adina E Croitoru
- Oncology Department, Fundeni Clinical Institute, 022238 Bucharest, Romania
- Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania
| | - Speranta M Iacob
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, 022238 Bucharest, Romania
| | | | | | - Cristian Gheorghe
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, 022238 Bucharest, Romania
| |
Collapse
|
8
|
Chaigneau T, Aguilera Munoz L, Oger C, Gourdeau C, Hentic O, Laurent L, Muller N, Dioguardi Burgio M, Gagaille MP, Lévy P, Rebours V, Hammel P, de Mestier L. Efficacy and tolerance of LV5FU2-carboplatin chemotherapy in patients with advanced pancreatic ductal adenocarcinoma after failure of standard regimens. Ther Adv Med Oncol 2023; 15:17588359231163776. [PMID: 37007630 PMCID: PMC10052496 DOI: 10.1177/17588359231163776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/24/2023] [Indexed: 03/30/2023] Open
Abstract
Background: Chemotherapy options in patients with advanced pancreatic ductal adenocarcinoma (PDAC) after failure of standard chemotherapies are limited. Objectives: We aimed to report the efficacy and safety of the leucovorin and 5-fluorouracil (LV5FU2) and carboplatin combination in this setting. Design: We performed a retrospective study including consecutive patients with advanced PDAC who received LV5FU2–carboplatin between 2009 and 2021 in an expert center. Methods: We measured overall survival (OS) and progression-free survival (PFS), and explored associated factors using Cox proportional hazard models. Results: In all, 91 patients were included (55% male, median age 62), with a performance status of 0/1 in 74% of cases. LV5FU2–carboplatin was mainly used in third (59.3%) or fourth line (23.1%), with three (interquartile range: 2.0–6.0) cycles administered on average. The clinical benefit rate was 25.2%. Median PFS was 2.7 months (95% CI: 2.4–3.0). At multivariable analysis, no extrahepatic metastases (p = 0.083), no ascites or opioid-requiring pain (p = 0.023), <2 prior treatment lines (p < 0.001), full dose of carboplatin (p = 0.004), and treatment initiation >18 months after initial diagnosis (p < 0.001) were associated with longer PFS. Median OS was 4.2 months (95% CI: 3.48–4.92) and was influenced by the presence of extrahepatic metastases (p = 0.058), opioid-requiring pain or ascites (p = 0.039), and number of prior treatment lines (0.065). Prior tumor response under oxaliplatin did not impact either PFS or OS. Worsening of preexisting residual neurotoxicity was infrequent (13.2%). The most common grade 3–4 adverse events were neutropenia (24.7%) and thrombocytopenia (11.8%). Conclusion: Although the efficacy of LV5FU2–carboplatin appears limited in patients with pretreated advanced PDAC, it may be beneficial in selected patients.
Collapse
Affiliation(s)
- Thomas Chaigneau
- Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
- Department of Hepatology, Gastroenterology, and Nutrition, Caen-Normandie University Hospital, Caen, France
| | - Lina Aguilera Munoz
- Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Caroline Oger
- Université Paris-Cité, Department of Pharmacy and Chemotherapy, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Clémence Gourdeau
- Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
- Department of Hepatology and Gastroenterology, Rouen University Hospital, Rouen, France
| | - Olivia Hentic
- Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Lucie Laurent
- Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Nelly Muller
- Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Marco Dioguardi Burgio
- Université Paris-Cité, Department of Radiology, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Marie-Pauline Gagaille
- Université Paris-Cité, Department of Pharmacy and Chemotherapy, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Philippe Lévy
- Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Vinciane Rebours
- Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Pascal Hammel
- Université Paris-Cité, Department of Digestive and Medical Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
- Université Paris-Saclay, Department of Digestive and Medical Oncology, Paul-Brousse Hospital (APHP.Sud), Villejuif, France
| | | |
Collapse
|
9
|
Möhring C, Frontado Graffe FJ, Bartels A, Sadeghlar F, Zhou T, Mahn R, Marinova M, Feldmann G, Brossart P, Glowka TR, Kalff JC, Strassburg CP, Gonzalez-Carmona MA. Second-line and third-line therapy with nanoliposomal irinotecan (nal-IRI) in pancreatic cancer: a single-center experience and review of literature. J Gastrointest Oncol 2023; 14:352-365. [PMID: 36915455 PMCID: PMC10007927 DOI: 10.21037/jgo-22-632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/07/2022] [Indexed: 02/17/2023] Open
Abstract
Background Prognosis of patients with pancreatic cancer is still extremely poor. First-line palliative therapies with FOLFIRINOX or gemcitabine/nab-paclitaxel have been established in the last decade. In the second-line, 5-FU/LV in combination with nanoliposomal irinotecan (nal-IRI) after gemcitabine has been shown to be effective. However, the use of nal-IRI as third-line therapy after FOLFIRINOX and gemcitabine-based chemotherapies is still controversial. In this study, we report about the use of 5-FU/LV + nal-IRI in a daily practice and analyze whether nal-IRI is an option as third-line therapy after FOLFIRINOX and gemcitabine/nab-paclitaxel. Methods This is a single center retrospective analysis of patients with irresectable pancreatic cancer who were treated with 5-FU/LV and nal-IRI from 2017 to 2021 as second- or third-line palliative treatment. Overall survival (OS), progression-free survival (PFS) and toxicity were analyzed, and multivariate analysis was used to identify independent prognostic factors. Results Twenty-nine patients receiving 5-FU/LV and nal-IRI were included in the analysis. The majority of patients (n=19) received 5-FU/nal-IRI as third-line therapy after pre-exposition to FOLFIRINOX and gemcitabine/nab-paclitaxel. Median OS and PFS were 9.33 months (95% CI: 3.37, 15.30) and 2.90 months (95% CI: 1.64, 4.16), respectively. Furthermore, patients receiving nal-IRI + 5-FU/LV as third-line treatment also showed some benefits, with no OS difference compared to second-line patients (9.33 vs. 10.27 months; HR: 1.85; 95% CI: 0.64, 5.41; P=0.253). Adverse effects were similar to reported trials. Conclusions In our study, the use of 5-FU/nal-IRI in unselected patients with advanced pancreatic cancer showed similar OS, PFS and tolerance as randomized prospective phase II/III trials. Interestingly, the use of 5-FU/nal-IRI seemed to be beneficial in third-line therapy, despite a pre-exposure to non-liposomal irinotecan.
Collapse
Affiliation(s)
- Christian Möhring
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | | | - Alexandra Bartels
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Farsaneh Sadeghlar
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Taotao Zhou
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Robert Mahn
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Milka Marinova
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | - Georg Feldmann
- Department of Internal Medicine III, University Hospital Bonn, Bonn, Germany
| | - Peter Brossart
- Department of Internal Medicine III, University Hospital Bonn, Bonn, Germany
| | - Tim R Glowka
- Department of Visceral Surgery, University Hospital Bonn, Bonn, Germany
| | - Jörg C Kalff
- Department of Visceral Surgery, University Hospital Bonn, Bonn, Germany
| | | | | |
Collapse
|
10
|
Testoni SGG, Petrone MC, Reni M, Di Serio C, Rancoita PM, Rossi G, Balzano G, Linzenbold W, Enderle M, Della-Torre E, De Cobelli F, Falconi M, Capurso G, Arcidiacono PG. EUS-guided ablation with the HybridTherm Probe as second-line treatment in patients with locally advanced pancreatic ductal adenocarcinoma: A case-control study. Endosc Ultrasound 2022; 11:383-392. [PMID: 36255026 PMCID: PMC9688129 DOI: 10.4103/eus-d-21-00200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/24/2022] [Indexed: 12/14/2022] Open
Abstract
Background and Objectives Data on the clinical efficacy of EUS-guided ablation using the HybridTherm-Probe (EUS-HTP) in locally advanced pancreatic ductal adenocarcinoma (LA-PDAC) are lacking. The aim of the study was to assess the impact of EUS-HTP added to chemotherapy (CT) on overall survival (OS) and progression-free survival (PFS) of LA-PDAC patients with local disease progression (DP) after first-line therapy, compared to CT alone in controls. Methods LA-PDAC cases, prospectively treated by EUS-HTP, were retrospectively compared to matched controls (1:2) receiving standard treatment. Study endpoints were the OS and PFS from local DP after first-line therapy, compared through log-rank test calculating hazard ratios and differences in restricted mean OS/PFS time (RMOST/RMPFST) within prespecified time points (4, 6, and 12 months). Results Thirteen cases and 26 controls were included. Clinical, tumor, and therapy features before and after first-line therapy were case-control balanced. The median OS and PFS were not significantly improved in cases over controls (months: 7 vs. 5 and 5 vs. 3, respectively). At 4 and 6 months, the RMPFST difference was in favor of cases (P = 0.0001 and P = 0.003, respectively). In cases and controls not candidate to further CT (N = 5 and N = 9), the median OS and PFS were not significantly improved in cases over controls (months: 6 vs. 3 and 4 vs. 2, respectively), but the RMPFST difference was in favor of cases at 4 months (P = 0.002). Conclusions In locally progressive PDAC patients experiencing failure of first-line therapy, EUS-HTP achieves a significantly better RMPFST up to 6 months compared to standard treatment, although without a significant impact on OS.
Collapse
Affiliation(s)
- Sabrina Gloria Giulia Testoni
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Maria Chiara Petrone
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Michele Reni
- Department of Oncology, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Clelia Di Serio
- Vita-Salute San Raffaele University, CUSSB, University Centre for Statistics in the Biomedical Sciences, Milan, Italy
| | - Paola Maria Rancoita
- Vita-Salute San Raffaele University, CUSSB, University Centre for Statistics in the Biomedical Sciences, Milan, Italy
| | - Gemma Rossi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Gianpaolo Balzano
- Department of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | | | | | - Emanuel Della-Torre
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Francesco De Cobelli
- Department of Radiology and Center for Experimental Imaging, Pancreas Translational and Clinical Research Center, Vita-Salute San Raffaele University, San Raffaele Hospital, Milan, Italy
| | - Massimo Falconi
- Department of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, Vita-Salute San Raffaele University, San Raffaele Hospital, Milan, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, Vita-Salute San Raffaele University, San Raffaele Hospital, Milan, Italy
| |
Collapse
|
11
|
Hemminki A, Heiniö C. Oncolytic adenovirus for treatment of malignant ascites. Mol Ther Oncolytics 2022; 26:302-303. [PMID: 36032634 PMCID: PMC9399406 DOI: 10.1016/j.omto.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics, Ltd., Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Corresponding author Akseli Hemminki, Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Corresponding author Camilla Heiniö, Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
12
|
Mohammed AA, Al-Zahrani O, Elsayed FM. The application of the Glasgow prognostic score to predict the survival in patients with metastatic pancreatic carcinoma. Indian J Palliat Care 2022; 28:406-412. [DOI: 10.25259/ijpc_81_2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 06/22/2022] [Indexed: 11/24/2022] Open
Abstract
Objectives:
Thither is a more pressing effort to think about chemotherapy (CTx) in second-line and beyond in patients with metastatic pancreatic cancer (mPC). The current work aimed to evaluate the value of the Glasgow prognostic score (GPS) and modified Glasgow prognostic score (mGPS) to predict the survival in patients receiving second-line CTx protocol.
Material and Methods:
We retrospectively reviewed the patients’ medical files with mPC who received second-line CTx protocol between September 2013 and December 2017. The GPS/mGPS graded from 0 to 2 based on C-reactive protein and serum albumin.
Results:
One hundred and sixty-nine patients with mPC were eligible. Survival of patients with Score 0 (GPS/mGPS) was better than that of Score 1 (GPS/mGPS) or Score 2 (GPS/mGPS), which was statistically significant (P < 0.001). Of 78 patients who died, only 16 patients belonged to Score 0 (GPS/mGPS), compared to 30 patients belonged to Score 1 (GPS/mGPS) and 32 patients belonged to Score 2 (GPS/mGPS). Univariate analysis showed that high GPS/mGPS (P < 0.000) as well as poor Eastern Cooperative Oncology Group Performance Status (P < 0.000) and metastasis either to the liver (P < 0.01) or lung (P < 0.04) were linked with worse prognosis. A statistically significant association was detected between the two scores. Cohen’s Kappa coefficient (k) was 0.9, SD = 0.03; 95% CI (0.787–0.922; P < 0.001).
Conclusion:
Our data suggested that GPS/mGPS is an easy and applicable index that may be used in daily practice and may help in the prognostic stratification of mPC patients to avert overtreatment in frail patients and raise the best supportive treatment concept.
Collapse
Affiliation(s)
| | - Omar Al-Zahrani
- Oncology Center, King Salman Armed Forces Hospital, Tabuk, Saudi Arabia,
| | - Fifi Mostafa Elsayed
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine Suez Canal, Suez, Egypt,
| |
Collapse
|
13
|
Ramaswamy A, Srinivas S, Chaudhari V, Bhargava P, Bhandare M, Shrikhande SV, Ostwal V. Systemic therapy in pancreatic ductal adenocarcinomas (PDACs)-basis and current status. Ecancermedicalscience 2022; 16:1367. [PMID: 35685956 PMCID: PMC9085164 DOI: 10.3332/ecancer.2022.1367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Indexed: 11/06/2022] Open
Abstract
A major shift in the approach to the management of pancreatic ductal adenocarcinomas (PDACs) has been the recognition of the systemic nature of the disease even in clinically and radiologically limited disease stages. The recalcitrant nature of PDAC is intrinsically related to the lack of therapeutic targets and dense surrounding stroma that limits the activity of currently available chemotherapeutic options. However, research is increasingly focusing on intensifying systemic management options in PDAC, resulting in gradual improvements in survival. Currently effective chemotherapeutic regimens like modified 5-fluorouracil-leucovorin-irinotecan-oxaliplatin and gemcitabine-nab-paclitaxel have improved outcomes in resectable and advanced PDAC. An increasing use of these regimens has also resulted in greater conversion of borderline resectable and locally advanced cancers to resection, though the most effective approach in this subgroup is yet to be identified. The current review presents an outline of the basic systemic nature of PDAC and current options of systemic therapy, predominantly chemotherapy .
Collapse
Affiliation(s)
- Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Dr E Borges Road, Parel, Mumbai 400012, India
| | - Sujay Srinivas
- Department of Medical Oncology, Tata Memorial Hospital, Dr E Borges Road, Parel, Mumbai 400012, India
| | - Vikram Chaudhari
- GI and HPB Services, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, India
| | - Prabhat Bhargava
- Department of Medical Oncology, Tata Memorial Hospital, Dr E Borges Road, Parel, Mumbai 400012, India
| | - Manish Bhandare
- GI and HPB Services, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, India
| | - Shailesh V Shrikhande
- GI and HPB Services, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, India
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Dr E Borges Road, Parel, Mumbai 400012, India
| |
Collapse
|
14
|
Ekström A, Brun E, Eberhard J, Segerlantz M. Second-line palliative chemotherapy, survival, and prognostic factors in patients with advanced pancreatic cancer. Acta Oncol 2021; 60:1580-1588. [PMID: 34486921 DOI: 10.1080/0284186x.2021.1973680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Pancreatic cancer is a highly lethal disease with a close association between incidence and mortality. First-line (FL) palliative chemotherapy prolongs survival and alleviates cancer-related symptoms. However, the survival benefit of second-line (SL) treatment is uncertain, as studies fail to consistently show prolonged survival for any given SL treatment, and in the absence of prognostic factors patients will receive a futile treatment. The aim of this study was to examine prognostic factors and survival in patients with pancreatic cancer, with special reference to SL therapy. MATERIAL AND METHODS This retrospective study included all patients with histopathologically verified pancreatic adenocarcinoma who received palliative chemotherapy at Skåne University Hospital and died between 1 Feb 2015 and 31 Dec 2017. RESULTS During the study period, a total of 170 patients with pancreatic cancer died after receiving palliative chemotherapy. Of these, 72 had received SL treatment after progression on FL treatment. Median overall survival (OS) from the start of SL treatment was 5.0 months (95% CI: 4.0-6.1). Median OS was 2.9 months for patients with performance status 2 at start of SL treatment compared to 5.3 months for patients with performance status 0-1 (p = .03), and 3.5 months (95% CI: 3.0-5.4) in patients with hypoalbuminemia (<36 g/L) at the start of SL therapy compared to 8.0 months (95% CI: 5.3-11.1) for patients with normal albumin levels (p = .009). Weight loss during FL therapy, a doubling of CA 19-9 after FL therapy, and length of progression-free survival during FL treatment were not associated with survival following SL therapy. CONCLUSION Poor performance status and hypoalbuminemia are negative prognostic factors for survival on SL palliative treatment in patients with advanced pancreatic cancer. Possible gain in survival should be carefully considered before initiating SL chemotherapy.
Collapse
Affiliation(s)
- Anders Ekström
- Department of Clinical Sciences, Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Oncology, Blekinge Hospital, Karlskrona, Sweden
| | - Eva Brun
- Department of Clinical Sciences, Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Jakob Eberhard
- Department of Clinical Sciences, Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Mikael Segerlantz
- Department of Clinical Sciences Lund, Oncology and Pathology, Institute for Palliative Care, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Palliative Care and Advanced Home Health Care, Primary Health Care Skåne, Region Skåne, Lund, Sweden
| |
Collapse
|
15
|
Chiu TJ, Su YY, Yang SH, Li CP, Bai LY, Chiang NJ, Chuang SC, Shan YS, Chan DC, Chen LT, Yen CJ, Peng CM, Chen YY, Chen JS, Chou WC. Liposomal irinotecan pre-emptive dose reduction in patients with pancreatic ductal adenocarcinoma: 667 patients' experience within a population-based study. Ther Adv Med Oncol 2021; 13:17588359211058255. [PMID: 34819998 PMCID: PMC8606735 DOI: 10.1177/17588359211058255] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/20/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Liposomal irinotecan (nal-IRI) plus 5-fluorouracil and leucovorin (5-FU/LV) is currently the standard second-line treatment for patients with pancreatic ductal adenocarcinoma (PDAC) after previous failed gemcitabine-based therapy. This population-based study aimed to evaluate the efficacy and safety of nal-IRI + 5-FU/LV and the association of pre-emptive nal-IRI dosing with treatment outcomes in patients with PDAC. Methods: We retrospectively enrolled a total of 667 consecutive patients with PDAC who received nal-IRI plus 5-FU/LV treatment between August 2018 and November 2020 at 9 medical centers in Taiwan. Patients were allocated into groups according to pre-emptive nal-IRI dosing (⩾75%, 50–74%, <50%) for comparison of treatment efficacy and safety. Results: The median overall survival (OS) and time to treatment failure (TTF) were 5.9 months [95% confidence interval (CI), 5.3–6.5] and 2.8 months (95% CI, 2.6–3.0), respectively. The median OS was 6.5 months (95% CI, 5.7–6.7), 5.0 months (95% CI, 3.4–6.5), and 4.1 months (95% CI, 2.7–5.6), respectively, among the ⩾75%, 50–74%, and <50% pre-emptive nal-IRI dosing groups, whereas the median TTF of the three groups was 3.0 months (95% CI, 2.6–3.4), 2.6 months (95% CI, 2.3–2.9), and 1.9 months (95% CI, 1.6–2.2), respectively. Pre-emptive nal-IRI dosing <50% was an independent negative prognostic factor for OS and TTF in multivariate analyses. The most common severe adverse events were neutropenia (22.9%), anemia (21.1%), and hypokalemia (15.4%). Patients in the <50% pre-emptive nal-IRI dosing group had a significantly lower incidence of neutropenia and non-neutropenic infection than those in the other groups. Conclusion: Our results support the use of nal-IRI + 5-FU/LV as standard clinical practice for treating patients with PDAC based on this large population-based study. Our findings encourage physicians to provide adequate doses of nal-IRI in order to achieve better outcomes without compromising safety profiles.
Collapse
Affiliation(s)
- Tai-Jan Chiu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung
| | - Yung-Yeh Su
- National Institute of Cancer Research, National Health Research Institutes, Tainan
| | - Shih-Hung Yang
- Department of Oncology, National Taiwan University Hospital, National Taiwan University, Taipei
| | - Chung-Pin Li
- Division of Gastroenterology and Hepatology, Department of Medicine and Division of Clinical Skills Training, Taipei
| | - Li-Yuan Bai
- Division of Hematology-Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung
| | - Nai-Jung Chiang
- National Institute of Cancer Research, National Health Research Institutes, Tainan
| | - Shih-Chang Chuang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung
| | - Yan-Shen Shan
- Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan
| | - De-Chuan Chan
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan
| | - Chia-Jui Yen
- Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Cheng-Ming Peng
- Department of Surgery, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung
| | - Yen-Yang Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung
| | - Jen-Shi Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan
| | - Wen-Chi Chou
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, 5 Fu-Hsing Street, 333 Kwei-Shan Shiang, Taoyuan
| |
Collapse
|
16
|
Manrai M, Tilak TVSVGK, Dawra S, Srivastava S, Singh A. Current and emerging therapeutic strategies in pancreatic cancer: Challenges and opportunities. World J Gastroenterol 2021; 27:6572-6589. [PMID: 34754153 PMCID: PMC8554408 DOI: 10.3748/wjg.v27.i39.6572] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/09/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic carcinoma (PC) is one of the leading causes of cancer-related deaths worldwide. Despite early detection and advances in therapeutics, the prognosis remains dismal. The outcome and therapeutic approach are dependent on the stage of PC at the time of diagnosis. The standard of care is surgery, followed by adjuvant chemotherapy. The advent of newer drugs has changed the landscape of adjuvant therapy. Moreover, recent trials have highlighted the role of neoadjuvant therapy and chemoradiotherapy for resectable and borderline resectable PC. As we progress towards a better understanding of tumor biology, genetics, and microenvironment, novel therapeutic strategies and targeted agents are now on the horizon. We have described the current and emerging therapeutic strategies in PC.
Collapse
Affiliation(s)
- Manish Manrai
- Department of Internal Medicine, Armed Forces Medical College, Pune 411040, Maharashtra, India
| | - T V S V G K Tilak
- Department of Internal Medicine, Armed Forces Medical College, Pune 411040, Maharashtra, India
| | - Saurabh Dawra
- Department of Internal Medicine, Command Hospital, Pune 411040, Maharashtra, India
| | - Sharad Srivastava
- Department of Internal Medicine, Command Hospital, Pune 411040, Maharashtra, India
| | - Anupam Singh
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Chandigarh 160011, India
| |
Collapse
|
17
|
A Prospective Feasibility Trial to Challenge Patient-Derived Pancreatic Cancer Organoids in Predicting Treatment Response. Cancers (Basel) 2021; 13:cancers13112539. [PMID: 34064221 PMCID: PMC8196829 DOI: 10.3390/cancers13112539] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 12/27/2022] Open
Abstract
Real-time isolation, propagation, and pharmacotyping of patient-derived pancreatic cancer organoids (PDOs) may enable treatment response prediction and personalization of pancreatic cancer (PC) therapy. In our methodology, PDOs are isolated from 54 patients with suspected or confirmed PC in the framework of a prospective feasibility trial. The drug response of single agents is determined by a viability assay. Areas under the curves (AUC) are clustered for each drug, and a prediction score is developed for combined regimens. Pharmacotyping profiles are obtained from 28 PDOs (efficacy 63.6%) after a median of 53 days (range 21-126 days). PDOs exhibit heterogeneous responses to the standard-of-care drugs, and are classified into high, intermediate, or low responder categories. Our developed prediction model allows a successful response prediction in treatment-naïve patients with an accuracy of 91.1% for first-line and 80.0% for second-line regimens, respectively. The power of prediction declines in pretreated patients (accuracy 40.0%), particularly with more than one prior line of chemotherapy. Progression-free survival (PFS) is significantly longer in previously treatment-naïve patients receiving a predicted tumor sensitive compared to a predicted tumor resistant regimen (mPFS 141 vs. 46 days; p = 0.0048). In conclusion, generation and pharmacotyping of PDOs is feasible in clinical routine and may provide substantial benefit.
Collapse
|
18
|
Gutierrez-Sainz L, Viñal D, Villamayor J, Martinez-Perez D, Garcia-Cuesta JA, Ghanem I, Custodio A, Feliu J. Prognostic factors in advanced pancreatic ductal adenocarcinoma patients-receiving second-line treatment: a single institution experience. Clin Transl Oncol 2021; 23:1838-1846. [PMID: 33866520 DOI: 10.1007/s12094-021-02589-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/08/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Second-line (2L) treatments for advanced pancreatic ductal adenocarcinoma (PDAC) achieve a modest benefit at the expense of potential toxicity. In the absence of predictive factors of response, the identification of prognostic factors could help in the therapeutic decisions-making. The purpose of this study was to assess the prognostic factors associated with shorter survival in patients with advanced PDAC who received 2L treatment. METHODS We conducted a single institution retrospective study, which included all patients with advanced PDAC who received 2L treatment between September 2006 and February 2020 at La Paz University Hospital, Madrid (Spain). Significant variables in the logistic regression model were used to create a prognostic score. RESULTS We included 108 patients. The median overall survival (OS) was 5.10 months (95%CI 4.02-6.17). In the multivariate analysis, time to progression (TTP) shorter than 4 months after first-line treatment (OR 4.53 [95%CI 1.28-16.00] p = 0.01), neutrophil-to-lymphocyte ratio (NLR) greater than 3 at the beginning of 2L (OR 9.07 [95%CI 1.82-45.16] p = 0.01) and CA-19.9 level higher than the upper limit of normal at the beginning of 2L (OR 7.83 [95%CI 1.30-49.97] p = 0.02) were independently associated with OS shorter than 3 months. The prognostic score classified patients into three prognostic groups (good, intermediate and poor) with significant differences in OS (p < 0.001). CONCLUSIONS TTP shorter than 4 months after first-line treatment, NLR greater than 3 and CA-19.9 level higher than the upper limit of normal at the beginning of 2L were associated with shorter overall survival. We developed a prognostic score that classifies patients with advanced PDAC into three prognostic groups after progression to the first-line. This score could help in the decision-making for 2L treatment.
Collapse
Affiliation(s)
- L Gutierrez-Sainz
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.
| | - D Viñal
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - J Villamayor
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - D Martinez-Perez
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - J A Garcia-Cuesta
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - I Ghanem
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - A Custodio
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.,Cátedra UAM-AMGEN, Madrid, Spain.,CIBERONC, Madrid, Spain
| | - J Feliu
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.,Cátedra UAM-AMGEN, Madrid, Spain.,CIBERONC, Madrid, Spain
| |
Collapse
|
19
|
Catalano M, Roviello G, Conca R, D'Angelo A, Palmieri VE, Panella B, Petrioli R, Ianza A, Nobili S, Mini E, Ramello M. Clinical Outcomes and Safety of Patients Treated with NAb-Paclitaxel Plus Gemcitabine in Metastatic Pancreatic Cancer: The NAPA Study. Curr Cancer Drug Targets 2020; 20:887-895. [PMID: 32957885 DOI: 10.2174/1568009620999200918122426] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/10/2020] [Accepted: 08/20/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND The phase III MPACT trial demonstrated the superiority of gemcitabine (Gem) combined with Nab-paclitaxel (Nab-P) versus gemcitabine alone in previously untreated patients with metastatic pancreatic ductal adenocarcinoma (PDAC). The purpose of this study was to evaluate the effect of Gem/Nab-P in routine clinical practice. METHODS From January 2015 to December 2018, patients with metastatic PDAC receiving firstline treatment with a combination of gemcitabine and Nab-paclitaxel were included in a multicentre retrospective observational study. Exploratory analyses of efficacy, and prognostic and predictive markers, were performed. RESULTS The cohort comprised 115 patients (median age 65 [range 50-84] years) with good performance status (ECOG PS 0-1). The median overall survival (OS) was 11 months (95% CI; 9-13) and the median progression-free survival (PFS) was 6 months (95% CI 5-7). Partial response and stable disease were achieved in 44 and 30 patients, respectively, yielding an overall disease control rate (DCR) of 64.3%. Grade 3-4 hematological toxicity frequency was 22.61% for neutropenia, 5.22% for anemia, and 3.48% for thrombocytopenia. Grade 3 asthenia was recorded in 2.61% of patients. No grade 4 non-hematological events were reported. Dose reduction was necessary in 51.3% of the patients. CONCLUSION Our results confirm the efficacy and safety of a first-line regimen comprising gemcitabine and Nab-paclitaxel in metastatic PDAC in a real-life population.
Collapse
Affiliation(s)
- Martina Catalano
- School of Human Health Sciences, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy
| | - Raffaele Conca
- Division of Medical Oncology, Department of Onco-Hematology, IRCCS-CROB, Referral Cancer Center of Basilicata, via Padre Pio 1, 85028 Rionero, Vulture (PZ), Italy
| | - Alberto D'Angelo
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Valeria Emma Palmieri
- School of Human Health Sciences, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Benedetta Panella
- School of Human Health Sciences, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Roberto Petrioli
- Department of Medicine, Surgery and Neurosciences, Medical Oncology Unit, University of Siena, Viale Bracci - Policlinico "Le Scotte" 53100, Siena, Italy
| | - Anna Ianza
- Oncology Unit, Department of Medical, Surgical, & Health Sciences, University of Trieste, Piazza Ospitale, Trieste, Italy
| | - Stefania Nobili
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy
| | - Enrico Mini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, viale Pieraccini, 6, 50139, Florence, Italy
| | - Monica Ramello
- 6Oncology Unit, Department of Medical, Surgical, & Health Sciences, University of Trieste, Piazza Ospitale, Trieste, Italy
| |
Collapse
|
20
|
Hsu CC, Liu KH, Chang PH, Chen PT, Hung CY, Hsueh SW, Yeh KY, Chen YY, Lu CH, Hung YS, Chou WC. Development and validation of a prognostic nomogram to predict survival in patients with advanced pancreatic cancer receiving second-line palliative chemotherapy. J Gastroenterol Hepatol 2020; 35:1694-1703. [PMID: 31711261 DOI: 10.1111/jgh.14926] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/31/2019] [Accepted: 11/02/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIM Given that a wide variation in tumor response rates and survival times suggests heterogeneity among the patients with advanced pancreatic cancer (APC) who underwent second-line (L2) chemotherapy, it is a challenge in clinical practice to identify patients who will receive the most benefit from L2 treatment. METHODS We selected 183 APC patients who received L2 palliative chemotherapy between 2010 and 2016 from a medical center as the development cohort. A Cox proportional hazard model was used to identify the prognostic factors and construct the nomogram. An independent cohort of 166 patients from three other hospitals was selected for external validation. RESULTS The nomogram was based on eight independent prognostic factors from the multivariate Cox model: sex, Eastern Cooperative Oncology Group performance status, reason for first-line treatment discontinuation, duration of first-line treatment, neutrophil-to-lymphocyte ratio, tumor stage, body mass index, and serum carbohydrate antigen 19-9 levels at the beginning of L2 treatment. The model exhibited good discrimination ability, with a C-index of 0.733 (95% confidence interval, 0.681-0.785) and 0.724 (95% confidence interval, 0.661-0.787) in the development and validation cohorts, respectively. The calibration plots of the development and validation cohorts showed optimal agreement between model prediction and actual observation in predicting survival probability at 6 months, 1 year, and 2 years. CONCLUSIONS This study developed and externally validated a prognostic model that accurately predicts the survival outcome of APC patients before L2 palliative chemotherapy, which could assist in clinical decision-making, counseling for treatment, and most importantly, prognostic stratification of patients.
Collapse
Affiliation(s)
- Chih-Chung Hsu
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Keng-Hao Liu
- Department of Surgery, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Pei-Hung Chang
- Department of Oncology, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan
| | - Ping-Tsung Chen
- Department of Oncology, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| | - Chia-Yen Hung
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Hema-oncology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Shun-Wen Hsueh
- Department of Oncology, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan
| | - Kun-Yun Yeh
- Department of Oncology, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan
| | - Yen-Yang Chen
- Department of Oncology, Chang Gung Memorial Hospital at Kaohsiung, Kaohsiung, Taiwan
| | - Chang-Hsien Lu
- Department of Oncology, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| | - Yu-Shin Hung
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Chi Chou
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
21
|
De Jesus-Acosta A, Narang A, Mauro L, Herman J, Jaffee EM, Laheru DA. Carcinoma of the Pancreas. ABELOFF'S CLINICAL ONCOLOGY 2020:1342-1360.e7. [DOI: 10.1016/b978-0-323-47674-4.00078-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
22
|
Lambert A, Schwarz L, Borbath I, Henry A, Van Laethem JL, Malka D, Ducreux M, Conroy T. An update on treatment options for pancreatic adenocarcinoma. Ther Adv Med Oncol 2019; 11:1758835919875568. [PMID: 31598142 PMCID: PMC6763942 DOI: 10.1177/1758835919875568] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 08/19/2019] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is one of the most lethal solid organ tumors. Due to the rising incidence, late diagnosis, and limited treatment options, it is expected to be the second leading cause of cancer deaths in high income countries in the next decade. The multidisciplinary treatment of this disease depends on the stage of cancer at diagnosis (resectable, borderline, locally advanced, and metastatic disease), and combines surgery, chemotherapy, chemoradiotherapy, and supportive care. The landscape of multidisciplinary pancreatic cancer treatment is changing rapidly, especially in locally advanced disease, and the number of treatment options in metastatic disease, including personalized medicine, innovative targets, immunotherapy, therapeutic vaccines, adoptive T-cell transfer, or stemness inhibitors, will probably expand in the near future. This review summarizes the current literature and provides an overview of how new therapies or new therapeutic strategies (neoadjuvant therapies, conversion surgery) will guide multidisciplinary disease management, future clinical trials, and, hopefully, will increase overall survival.
Collapse
Affiliation(s)
- Aurélien Lambert
- Department of Medical Oncology, Institut de Cancérologie de Lorraine and Université de Lorraine, Nancy, France
| | - Lilian Schwarz
- Department of Digestive Surgery, Rouen University Hospital and Université de Rouen Normandie, France
| | - Ivan Borbath
- Department of Gastroenterology and Digestive Oncology, Cliniques Universitaires Saint-Luc and Université Catholique de Louvain, Brussels, Belgium
| | - Aline Henry
- Department of Supportive Care in Oncology, Institut de Cancérologie de Lorraine, Nancy, France
| | - Jean-Luc Van Laethem
- Department of Gastroenterology and Digestive Oncology, Erasme University Hospital, Université Libre de Bruxelles, Belgium
| | - David Malka
- Department of Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Michel Ducreux
- Department of Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Thierry Conroy
- Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 50519 Vandoeuvre-lès-Nancy CEDEX, France
| |
Collapse
|
23
|
Gemcitabine Combined with the mTOR Inhibitor Temsirolimus in Patients with Locally Advanced or Metastatic Pancreatic Cancer. A Hellenic Cooperative Oncology Group Phase I/II Study. Target Oncol 2019; 13:715-724. [PMID: 30488350 DOI: 10.1007/s11523-018-0605-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The prognosis of patients with advanced pancreatic cancer is dismal, and there is a need for novel and effective treatments. OBJECTIVES Tο determine the maximum tolerated dose (MTD) and dose-limiting toxicities (DLTs) of a novel gemcitabine (G) and temsirolimus (T) combination (phase I) and estimate the 6-month progression-free survival (PFS) in patients treated with the T + G combination (phase II). PATIENTS AND METHODS Eligible patients with histologically confirmed inoperable or metastatic pancreatic carcinoma (MPC) were entered into the trial. G was given bi-weekly and T weekly in a 4-week cycle. The first dose level was set at G 800 mg/m2 and T 10 mg. G was escalated in increments of 200 mg/m2 and T in increments of 5 mg until DLT was reached, and the recommended dose was used for the phase II part. RESULTS Thirty patients were enrolled in the phase I component at the pre-planned six dose levels; one bilirubin DLT of grade III occurred at the first dose level. The MTD was established as the approved doses of both drugs. Fifty-five patients were entered into the phase II component. Median relative dose intensities administered in the first cycle were 0.75 for T and 0.99 for G. Grade 3-4 hematological toxicities were recorded in 87.3% of patients. The most common non-hematological adverse events were metabolic disorders (81.8%) followed by gastrointestinal disorders (63.6%). Median PFS was 2.69 months (95% CI 1.74-4.95) and median OS was 4.95 months (95% CI 3.54-6.85), while the 6-month PFS rate was 30.9%. CONCLUSIONS Combination of G and T is feasible in patients with locally advanced or MPC with manageable side effects, but lacks clinical efficacy. The study was registered in the Australian New Zealand Clinical Trials Registry (ACTRN12611000643976).
Collapse
|
24
|
NAPOLI-1 phase 3 study of liposomal irinotecan in metastatic pancreatic cancer: Final overall survival analysis and characteristics of long-term survivors. Eur J Cancer 2019; 108:78-87. [PMID: 30654298 DOI: 10.1016/j.ejca.2018.12.007] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Liposomal irinotecan (nal-IRI) plus 5-fluorouracil and leucovorin (5-FU/LV) is approved for patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) previously treated with gemcitabine-based therapy. This approval was based on significantly improved median overall survival compared with 5-FU/LV alone (6.1 vs 4.2 months; hazard ratio [HR], 0.67) in the global phase 3 NAPOLI-1 trial. Here, we report the final survival analysis and baseline characteristics associated with long-term survivors (survival of ≥1 year) in the NAPOLI-1 trial. PATIENTS AND METHODS Patients with mPDAC were randomised to receive nal-IRI + 5-FU/LV (n = 117), nal-IRI (n = 151), or 5-FU/LV (n = 149) for the first 4 weeks of 6-week cycles. Baseline characteristics and efficacy in the overall population were compared with those in patients who survived ≥1 year. Through 16th November 2015, 382 overall survival events had occurred. RESULTS The overall survival advantage for nal-IRI+5-FU/LV vs 5-FU/LV was maintained from the original nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1) analysis (6.2 vs 4.2 months, respectively; HR, 0.75; 95% confidence interval: 0.57-0.99). Median progression-free survival, objective response rate and disease control rate also favoured nal-IRI+5-FU/LV therapy. Estimated one-year overall survival rates were 26% with nal-IRI+5-FU/LV and 16% with 5-FU/LV. Baseline characteristics associated with long-term survival in the nal-IRI+5-FU/LV arm were Karnofsky performance status ≥90, age ≤65 years, lower CA19-9 levels, neutrophil-to-lymphocyte ratio ≤5 and no liver metastases. No new safety concerns were detected. CONCLUSIONS The survival benefits of nal-IRI+5-FU/LV versus 5-FU/LV were maintained over an extended follow-up, and prognostic markers of survival ≥1 year were identified. CLINICAL TRIAL REGISTRATION NUMBER NCT01494506.
Collapse
|
25
|
Chen J, Chen L, Yu J, Xu Y, Wang X, Zeng Z, Liu N, Xu F, Yang S. Meta‑analysis of current chemotherapy regimens in advanced pancreatic cancer to prolong survival and reduce treatment‑associated toxicities. Mol Med Rep 2019; 19:477-489. [PMID: 30431091 PMCID: PMC6297739 DOI: 10.3892/mmr.2018.9638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 10/23/2018] [Indexed: 02/05/2023] Open
Abstract
Unresectable advanced pancreatic cancer (APC) is a highly lethal malignancy. Although numerous chemotherapeutic regimens are available, evidence regarding the survival extension, the life quality improvement, the associated risks and occurrence rates of adverse effects, is required. The effects of 19 chemotherapy regimens on survival and treatment‑associated toxicities in the context of APC treatment were comparatively assessed. A total of 23 randomized controlled trials were included in this network meta‑analysis. For overall survival, five regimens, Gemcitabine (Gem)+radiotherapy (Radio), Gem+cisplatin (Cis), Gem+erlotinib (Erl)+bevacizumab (Bev), Gem+capecitabine (Cap)+Erl, and Gem+exatecan, were the most effective treatments, according to their respective high surface under the cumulative ranking (SUCRA) probabilities. Regarding the progression‑free survival, five regimens, including Gem+Radio, Gem+Erl+Bev, Gem+Cis, Gem+Cap+Erl and Gem+pemetrexed, were the most effective treatments based on their SUCRA probabilities. Each regimen exhibited advantages and disadvantages, and 14 common treatment‑associated toxicities were present in different proportions. The three principal toxic effects included haematological, gastrointestinal and constitutional symptoms. To improve survival, chemotherapy regimens with high SUCRA probabilities require prioritizing. Although treatment‑associated toxicities are unavoidable, the regimens presented toxicities in distinct proportions. Therefore, clinicians should assess the disease status of the patients, and balance the benefits and risks of the selected treatment.
Collapse
Affiliation(s)
- Jie Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR 999077, P.R. China
- Department of Orthopedics, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Linli Chen
- Division of General Practice, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jianping Yu
- Department of Neurology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Yanmei Xu
- Division of General Practice, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaohui Wang
- Department of General Surgery, Bayingol Mongolia Autonomous Prefecture People's Hospital, Urumqi, Xinjiang Uygur Autonomous Region 841300, P.R. China
| | - Ziqian Zeng
- Public Health School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Ning Liu
- Department of Medicine, Sunshine Guojian Pharmaceutical Co., Ltd., Shanghai 201203, P.R. China
| | - Fan Xu
- Public Health School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- Interdisciplinary Division of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, SAR 999077, P.R. China
| | - Shu Yang
- Public Health School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| |
Collapse
|
26
|
Neuzillet C, Gaujoux S, Williet N, Bachet JB, Bauguion L, Colson Durand L, Conroy T, Dahan L, Gilabert M, Huguet F, Marthey L, Meilleroux J, de Mestier L, Napoléon B, Portales F, Sa Cunha A, Schwarz L, Taieb J, Chibaudel B, Bouché O, Hammel P. Pancreatic cancer: French clinical practice guidelines for diagnosis, treatment and follow-up (SNFGE, FFCD, GERCOR, UNICANCER, SFCD, SFED, SFRO, ACHBT, AFC). Dig Liver Dis 2018; 50:1257-1271. [PMID: 30219670 DOI: 10.1016/j.dld.2018.08.008] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND This document is a summary of the French intergroup guidelines regarding the management of pancreatic adenocarcinoma (PA), updated in July 2018. DESIGN This collaborative work was produced under the auspices of all French medical and surgical societies involved in the management of PA. It is based on the previous guidelines, recent literature review and expert opinions. Recommendations were graded in three categories, according to the level of evidence. RESULTS Over the last seven years, significant changes in PA management have been implemented in clinical practice. Imaging/staging: diffusion magnetic resonance imaging is useful before surgery to rule out small liver metastases. SURGERY centralization of pancreatic surgery in expert centers is associated with a decreased postoperative mortality. Adjuvant chemotherapy: modified FOLFIRINOX in fit patients, or gemcitabine, or 5-FU, or gemcitabine plus capecitabine, to be discussed on a case-by-case basis. Locally advanced PA: no survival benefit of chemoradiotherapy. Metastatic PA: FOLFIRINOX and gemcitabine plus nab-paclitaxel combination are first-line standards in fit patients; second-line with 5FU/nal-IRI or 5FU/oxaliplatin combination after first-line gemcitabine. CONCLUSION Guidelines for management of PA are continuously evolving and need to be regularly updated. This constant progress is made possible through clinical and translational research. However, as each individual case is particular, they cannot substitute to multidisciplinary tumor board discussion.
Collapse
Affiliation(s)
- Cindy Neuzillet
- Department of Medical Oncology, Curie Institute, Versailles Saint-Quentin University (UVSQ), Saint-Cloud, France.
| | - Sébastien Gaujoux
- Department of Digestive, Hepato-Biliary and Pancreatic Surgery, Cochin Hospital, AP-HP, Paris Descartes Faculty of Medicine, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Nicolas Williet
- Hepato-Gastroenterology Department, University Hospital of Saint-Etienne, Saint Priest en Jarez, France
| | - Jean-Baptiste Bachet
- Hepato-Gastroenterology Department, Pitié Salpétrière University Hospital, AP-HP, Paris Cedex 13, France
| | - Lucile Bauguion
- Hepato-Gastroenterology Department, Departmental Hospital Center, La Roche sur Yon, France
| | - Laurianne Colson Durand
- Department of Radiotherapy, Henri Mondor Hospital, AP-HP, Université Paris Est Creteil, Créteil, France
| | - Thierry Conroy
- Department of Medical Oncology, Lorraine Institute of Oncology and Lorraine University, Vandoeuvre-lès-Nancy Cedex, France
| | - Laetitia Dahan
- Digestive Oncology Department, "DACCORD" (Digestif, Anatomie pathologique, Chirurgie, CISIH, Oncologie, Radiothérapie, Dermatologie) pole, CHU Timone, Marseille Cedex 05, France
| | - Marine Gilabert
- Paoli Calmettes Institute, Department of Medical Oncology and Cancer Research Center of Marseille (CRCM), INSERM U1068 Stress Cell, Aix-Marseille University, Marseille, France
| | - Florence Huguet
- Department of Oncology and Radiotherapy, Tenon Hospital, East Paris University Hospitals, AP-HP, Paris Sorbonne University, Paris, France
| | - Lysiane Marthey
- Gastroenterology Department, Béclère Hospital, AP-HP, Clamart, France
| | - Julie Meilleroux
- Pathology Department, Toulouse University Hospital, Toulouse, France
| | - Louis de Mestier
- Department of Gastroenterology-Pancreatology, Beaujon Hospital, APHP, Paris 7 University, Clichy, France
| | - Bertrand Napoléon
- Jean Mermoz Private Hospital, Ramsay Générale de Santé, Lyon, France
| | - Fabienne Portales
- Digestive Oncology Department, Regional Institute of Cancer, Montpellier, France
| | - Antonio Sa Cunha
- INSERM UMR 935, Paul Brousse Hospital, Hepatobiliary Center, AP-HP, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Lilian Schwarz
- Department of Digestive Surgery, Hôpital Charles Nicolle, Rouen University Hospital, Rouen, France and Genomic and Personalized Medicine in Cancer and Neurological Disorders, UMR 1245 INSERM, Rouen University, France
| | - Julien Taieb
- Hepato-Gastroenterology and Digestive Oncology Department, Georges Pompidou European Hospital, AP-HP, Paris, France
| | - Benoist Chibaudel
- Department of Medical Oncology, Franco-British Institute, Levallois-Perret, France
| | - Olivier Bouché
- Hepato-Gastroenterology and Digestive Oncology Department, Robert Debré University Hospital, Avenue Général Koenig, 51092 Reims Cedex, France
| | - Pascal Hammel
- Department of Digestive Oncology, Beaujon University Hospital (AP-HP), Paris VII Diderot University, Clichy-la-Garenne, France.
| |
Collapse
|
27
|
Abstract
OPINION STATEMENT Managing patients with metastatic pancreatic adenocarcinoma (mPDA) is a challenging proposition for any treating oncologist. Although the potency of first-line therapies has improved with the approvals of FOLFIRINOX and gemcitabine plus nab-paclitaxel, many patients are unable to derive significant benefit from later lines of therapy upon progression. Enrollment on clinical trials remains among the best options for patients with mPDA in all lines of therapy. At our institution, we routinely check for microsatellite instability (MSI-H) and perform next-generation sequencing (NGS) at the time of diagnosis in all good performance status mPDA patients. Although MSI-H status is only found in 1% of patients with mPDA, given pembrolizumab's tissue-agnostic approval for MSI-H tumors in later-line settings, it is a viable option when deciding on subsequent lines of therapy. Any use of immune therapy in mPDA is investigational outside the MSI-H setting. NGS can identify BRCA or other DNA damage response (DDR) defects in patients which can predict sensitivity to platinum-based therapies and influence choice of both initial and later lines of therapy. It can also identify rare actionable genomic alterations such as HER2 (2%) and TRK fusions (0.1%) and offer patients the option of enrollment on clinical trials with agents targeting these or other identified alterations. We believe enrolling mPDA patients on clinical trials with immune-modulating agents is critical to determine if there are other patient subsets, outside of the MSI-H setting, who would benefit from these approaches. Immunotherapy's general tolerability and potential to generate durable responses make it particularly appealing for mPDA patients. Although single-modality immunotherapy such as checkpoint inhibitors or vaccines have not demonstrated efficacy in this disease, combinatorial strategies targeting unique aspects of PDA including the tumor microenvironment and desmoplastic stroma have shown preclinical or early-phase success. Validating these treatments with later-phase prospective studies is essential to making immunotherapy a routine component of the treatment armamentarium for mPDA patients.
Collapse
Affiliation(s)
- Satya Das
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN, 37232, USA.
| | - Jordan Berlin
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN, 37232, USA
| | - Dana Cardin
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN, 37232, USA
| |
Collapse
|
28
|
Hurwitz H, Van Cutsem E, Bendell J, Hidalgo M, Li CP, Salvo MG, Macarulla T, Sahai V, Sama A, Greeno E, Yu KH, Verslype C, Dawkins F, Walker C, Clark J, O'Reilly EM. Ruxolitinib + capecitabine in advanced/metastatic pancreatic cancer after disease progression/intolerance to first-line therapy: JANUS 1 and 2 randomized phase III studies. Invest New Drugs 2018; 36:683-695. [PMID: 29508247 PMCID: PMC6752723 DOI: 10.1007/s10637-018-0580-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/20/2018] [Indexed: 12/18/2022]
Abstract
Background Ruxolitinib, a Janus kinase 1 (JAK1)/JAK2 inhibitor, plus capecitabine improved overall survival (OS) vs capecitabine in a subgroup analysis of patients with metastatic pancreatic cancer and systemic inflammation (C-reactive protein [CRP] >13 mg/dL) in the randomized phase II RECAP study. We report results from two randomized phase III studies, JANUS 1 (NCT02117479) and JANUS 2 (NCT02119663). Patients and Methods Adults with advanced/metastatic pancreatic cancer, one prior chemotherapy regimen and CRP >10 mg/L were randomized 1:1 (stratified by modified Glasgow Prognostic Score [1 vs 2] and Eastern Cooperative Oncology Group performance status [0/1 vs 2]) to 21-day cycles of ruxolitinib 15 mg twice daily plus capecitabine 2000 mg/m2/day (Days 1-14) or placebo plus capecitabine. The primary endpoint was OS. Results Both studies were terminated following a planned interim futility/efficacy analysis of JANUS 1. Overall, 321 and 86 patients were randomized in JANUS 1 (ruxolitinib: n = 161; placebo: n = 160) and JANUS 2 (ruxolitinib: n = 43; placebo: n = 43). There was no significant difference in OS or progression-free survival (PFS) between treatments in JANUS 1 (OS: hazard ratio [HR], 0.969, 95% confidence interval [CI], 0.747-1.256; PFS: HR, 1.056; 95% CI, 0.827-1.348) or JANUS 2 (OS: HR, 1.584; 95% CI, 0.886-2.830; PFS: HR, 1.166; 95% CI, 0.687-1.978). The most common hematologic adverse event was anemia. No new safety signals with ruxolitinib or capecitabine were identified. Conclusions Ruxolitinib plus capecitabine was well tolerated in refractory pancreatic cancer patients; this combination did not improve survival.
Collapse
Affiliation(s)
- Herbert Hurwitz
- Duke University Medical Center, Campus mail 439 Seeley-mudd Bldg, 10 Bryan Searle Drive, Duke University M, Durham, NC, 27710, USA
| | - Eric Van Cutsem
- Clinical Digestive Oncology, University Hospitals Leuven and KU Leuven, UZ Herestraat 49, 3000, Leuven, Belgium
| | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, 250 25th Ave N, Nashville, TN, 37203, USA
| | - Manuel Hidalgo
- Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Chung-Pin Li
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Road, Beitou District, Taipei, 11217, Taiwan
- School of Medicine, National Yang-Ming University, No. 155, Sec. 2, Li-Nong Street, Beitou District, Taipei, 112, Taiwan
| | - Marcelo Garrido Salvo
- Pontificia Universidad Católica de Chile, Av Libertador Bernardo O'Higgins, 340, Santiago, Región Metropolitana, Chile
| | - Teresa Macarulla
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital (HUVH), Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Vaibhav Sahai
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Cancer Center Floor B1 Reception E, 1500 E Medical Center Dr SPC 5912, Ann Arbor, MI, 48109-5912, USA
| | - Ashwin Sama
- Thomas Jefferson University Hospital, 925 Chestnut Street, Suite 320A, Philadelphia, PA, 19107, USA
| | - Edward Greeno
- Division of Hematology, Oncology and Transplantation, University of Minnesota, 420 Delaware Street SE, MMC 480, Minneapolis, MN, 55455, USA
| | - Kenneth H Yu
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY, 10065, USA
| | - Chris Verslype
- Clinical Digestive Oncology, University Hospitals Leuven and KU Leuven, UZ Herestraat 49, 3000, Leuven, Belgium
| | - Fitzroy Dawkins
- Incyte Corporation, 1801 Augustine Cut-off, Wilmington, DE, 19803, USA
| | - Chris Walker
- Incyte Corporation, 1801 Augustine Cut-off, Wilmington, DE, 19803, USA
| | - Jason Clark
- Incyte Corporation, 1801 Augustine Cut-off, Wilmington, DE, 19803, USA
| | - Eileen M O'Reilly
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY, 10065, USA.
- Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
29
|
Targeting Intramembrane Protein-Protein Interactions: Novel Therapeutic Strategy of Millions Years Old. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 111:61-99. [PMID: 29459036 PMCID: PMC7102818 DOI: 10.1016/bs.apcsb.2017.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Intramembrane protein-protein interactions (PPIs) are involved in transmembrane signal transduction mediated by cell surface receptors and play an important role in health and disease. Recently, receptor-specific modulatory peptides rationally designed using a general platform of transmembrane signaling, the signaling chain homooligomerization (SCHOOL) model, have been proposed to therapeutically target these interactions in a variety of serious diseases with unmet needs including cancer, sepsis, arthritis, retinopathy, and thrombosis. These peptide drug candidates use ligand-independent mechanisms of action (SCHOOL mechanisms) and demonstrate potent efficacy in vitro and in vivo. Recent studies surprisingly revealed that in order to modify and/or escape the host immune response, human viruses use similar mechanisms and modulate cell surface receptors by targeting intramembrane PPIs in a ligand-independent manner. Here, I review these intriguing mechanistic similarities and discuss how the viral strategies optimized over a billion years of the coevolution of viruses and their hosts can help to revolutionize drug discovery science and develop new, disruptive therapies. Examples are given.
Collapse
|
30
|
Shen ZT, Sigalov AB. Novel TREM-1 Inhibitors Attenuate Tumor Growth and Prolong Survival in Experimental Pancreatic Cancer. Mol Pharm 2017; 14:4572-4582. [PMID: 29095622 DOI: 10.1021/acs.molpharmaceut.7b00711] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pancreatic cancer (PC) is a highly lethal cancer with an urgent need to expand the limited treatment options for patients. Tumor-associated macrophages (TAMs) promote tumor aggressiveness and metastasis. High expression of triggering receptor expressed on myeloid cells 1 (TREM-1) on TAMs directly correlates with poor survival in patients with non-small cell lung cancer (NSCLC). We have previously hypothesized that blockade of TREM-1 could be a promising therapeutic strategy to treat cancer and shown that the novel, ligand-independent TREM-1 inhibitory peptides rationally designed using the signaling chain homooligomerization (SCHOOL) strategy suppress NSCLC growth in vivo. Here, we evaluated the therapeutic potential of these inhibitors in three human PC xenograft mouse models. Administration of SCHOOL peptides resulted in a strong antitumor effect achieving an optimal treatment/control (T/C) value of 19% depending on the xenograft and formulation used and persisting even after treatment was halted. The effect correlated significantly with increased survival and suppressed TAM infiltration. The peptides were well-tolerated when deployed either in free form or formulated into lipopeptide complexes for peptide half-life extension and targeted delivery. Finally, blockade of TREM-1 significantly reduced serum levels of interleukin (IL)-1α, IL-6, and macrophage colony-stimulating factor (M-CSF), but not vascular endothelial growth factor, suggesting M-CSF-dependent antitumor mechanisms. Collectively, these promising data suggest that SCHOOL TREM-1-specific peptide inhibitors have a cancer type independent, therapeutically beneficial antitumor activity and can be potentially used as a stand-alone therapy or as a component of combinational therapy for PC, NSCLC, and other solid tumors.
Collapse
Affiliation(s)
- Zu T Shen
- SignaBlok, Inc. , P.O. Box 4064, Shrewsbury, Massachusetts 01545, United States
| | - Alexander B Sigalov
- SignaBlok, Inc. , P.O. Box 4064, Shrewsbury, Massachusetts 01545, United States
| |
Collapse
|
31
|
Clinical Management. Cancer J 2017; 23:355-361. [DOI: 10.1097/ppo.0000000000000294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Osipov A, Nissen N, Rutgers J, Dhall D, Naziri J, Chopra S, Li Q, Hendifar AE, Tuli R. Redefining the Positive Margin in Pancreatic Cancer: Impact on Patterns of Failure, Long-Term Survival and Adjuvant Therapy. Ann Surg Oncol 2017; 24:3674-3682. [PMID: 28871564 DOI: 10.1245/s10434-017-6076-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Indexed: 12/24/2022]
Abstract
PURPOSE There is debate regarding the definition and clinical significance of margin clearance in pancreatic ductal adenocarcinoma (PDA). A comprehensive archival analysis of surgical resection margins was performed to determine the effect on locoregional recurrence and survival, and the impact of adjuvant therapy in PDA. METHODS We identified 105 patients with resected PDA. Pancreatic, anterior, bile duct, and posterior surgical resection margins (PM; posterior surface, uncinate and vascular groove) were identified. Three pathologists reviewed all archival surgical specimens and recategorized each margin as tumor at ink/transected, <0.5, 0.5-1, >1-2, or >2 mm from the inked surface. The impact of these and other clinical variables was assessed on local control, disease-free survival (DFS), and overall survival (OS). RESULTS Among all margins, PM clearance up to 2 mm was prognostic of DFS (p = 0.01) and OS (p = 0.01). Dichotomizing the PM at 2 mm revealed it to be an independent predictor of local recurrence-free survival [hazard ratio HR] 0.20, 95% confidence interval [CI] 0.048-0.881, p = 0.033), DFS (HR 0.46, 95% CI 0.22-0.96, p = 0.03), and OS (HR 0.31, 95% CI 0.14-0.74, p = 0.008). A margin status of >2 mm was also prognostic of OS in patients who received adjuvant chemotherapy (HR 0.31, 95% CI 0.11-0.89, p = 0.03), however this difference was mitigated in patients receiving adjuvant chemoradiotherapy (HR 0.40, 95% CI 0.10-1.58, p = 0.19). CONCLUSION These data highlight the clinical significance of the PM and the lack of significance of other resection margins. Clearance in excess of 2 mm should be considered to improve long-term clinical outcomes. The use of adjuvant radiotherapy should be strongly considered in patients with PMs <2 mm.
Collapse
Affiliation(s)
- Arsen Osipov
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nicholas Nissen
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joanne Rutgers
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Deepti Dhall
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jason Naziri
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shefali Chopra
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Quanlin Li
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Richard Tuli
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
33
|
Kent OA, Sandi MJ, Rottapel R. Co-dependency between KRAS addiction and ARHGEF2 promotes an adaptive escape from MAPK pathway inhibition. Small GTPases 2017; 10:441-448. [PMID: 28656876 PMCID: PMC6748365 DOI: 10.1080/21541248.2017.1337545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oncogenic KRAS engages multiple effector pathways including the MAPK cascade to promote proliferation and survival of pancreatic cancer cells. KRAS-transformed cancer cells exhibit oncogene addiction to sustained activity of RAS for maintenance of malignant phenotypes. Previously, we have shown an essential role for the RHO guanine exchange factor ARHGEF2 for growth and survival of RAS-transformed pancreatic tumors. Here, we have determined that pancreatic cancer cells demonstrating KRAS addiction are significantly dependent on expression of ARHGEF2. Furthermore, enforced expression of ARHGEF2 desensitizes cells to pharmacological MEK inhibition and initiates a positive feedback loop which activates ERK phosphorylation and the downstream ARHGEF2 promoter. Therefore, targeting ARHGEF2 expression may increase the efficacy of MAPK inhibitors for treatment of RAS-dependent pancreatic cancers.
Collapse
Affiliation(s)
- Oliver A Kent
- Princess Margaret Cancer Centre, University Health Network, Toronto Medical Discovery Tower, University of Toronto , Toronto , Canada
| | - Maria-Jose Sandi
- Princess Margaret Cancer Centre, University Health Network, Toronto Medical Discovery Tower, University of Toronto , Toronto , Canada
| | - Robert Rottapel
- Princess Margaret Cancer Centre, University Health Network, Toronto Medical Discovery Tower, University of Toronto , Toronto , Canada.,Department of Medicine , Toronto , Canada.,Department of Medical Biophysics , Toronto , Canada.,Department of Immunology , Toronto , Canada.,Division of Rheumatology, St. Michael's Hospital , Toronto , Canada
| |
Collapse
|
34
|
Genomic Variations in Pancreatic Cancer and Potential Opportunities for Development of New Approaches for Diagnosis and Treatment. Int J Mol Sci 2017; 18:ijms18061201. [PMID: 28587243 PMCID: PMC5486024 DOI: 10.3390/ijms18061201] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 04/30/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Human pancreatic cancer has a very poor prognosis with an overall five-year survival rate of less than 5% and an average median survival time of six months. This is largely due to metastatic disease, which is already present in the majority of patients when diagnosed. Although our understanding of the molecular events underlying multi-step carcinogenesis in pancreatic cancer has steadily increased, translation into more effective therapeutic approaches has been inefficient in recent decades. Therefore, it is imperative that novel and targeted approaches are designed to facilitate the early detection and treatment of pancreatic cancer. Presently, there are numerous ongoing studies investigating the types of genomic variations in pancreatic cancer and their impact on tumor initiation and growth, as well as prognosis. This has led to the development of therapeutics to target these genetic variations for clinical benefit. Thus far, there have been minimal clinical successes directly targeting these genomic alterations; however research is ongoing to ultimately discover an innovative approach to tackle this devastating disease. This review will discuss the genomic variations in pancreatic cancer, and the resulting potential diagnostic and therapeutic implications.
Collapse
|
35
|
Grasso C, Jansen G, Giovannetti E. Drug resistance in pancreatic cancer: Impact of altered energy metabolism. Crit Rev Oncol Hematol 2017; 114:139-152. [PMID: 28477742 DOI: 10.1016/j.critrevonc.2017.03.026] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023] Open
|
36
|
Second-line chemotherapy for advanced pancreatic cancer: Which is the best option? Crit Rev Oncol Hematol 2017; 115:1-12. [PMID: 28602164 DOI: 10.1016/j.critrevonc.2017.03.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 02/28/2017] [Accepted: 03/21/2017] [Indexed: 12/17/2022] Open
Abstract
Despite recent biological insight and therapeutic advances, the prognosis of advanced pancreatic cancer still remains poor. For more than 15 years, gemcitabine monotherapy has been the cornerstone of first-line treatment. Recently, prospective randomized trials have shown that novel upfront combination regimens tested in prospective randomized trials have resulted in improved patients' outcome increasing the proportion of putative candidate to second-line therapy. There is no definite standard of care after disease progression. A novel formulation in which irinotecan is encapsulated into liposomal-based nanoparticles may increase the efficacy of the drug without incrementing its toxicity. NAPOLI-1 was the first randomized trial to compare nanoliposomal irinotecan and fluorouracil-leucovorin (5-FU/LV) to 5-FU/LV alone after a gemcitabine-based chemotherapy. This review focuses on the current data for the management of second-line treatment for metastatic pancreatic adenocarcinoma, presents the most interesting ongoing clinical trials and illustrates the biologically-driven future options beyond disease progression.
Collapse
|
37
|
Xiao L, Erb U, Zhao K, Hackert T, Zöller M. Efficacy of vaccination with tumor-exosome loaded dendritic cells combined with cytotoxic drug treatment in pancreatic cancer. Oncoimmunology 2017; 6:e1319044. [PMID: 28680753 DOI: 10.1080/2162402x.2017.1319044] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/04/2017] [Accepted: 04/07/2017] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer (PaCa) has a dismal prognosis and adjuvant immunotherapy frequently is of low efficacy due to immunosuppressive features of PaCa and PaCa-stroma. We here explored, whether the efficacy of vaccination with tumor-exosome (TEX)-loaded dendritic cells (DC) can be improved by combining with drugs affecting myeloid-derived suppressor cells (MDSC). Experiments were performed with the UNKC6141 PaCa line. UNKC6141 TEX-loaded DC were weekly intravenously injected, mice additionally receiving Gemcitabine (GEM) and/or ATRA and/or Sunitinib (Sun). UNKC6141 grow aggressively after subcutaneous and orthotopic application and are consistently recovered in peripheral blood, bone marrow, lung and frequently liver. Vaccination with DC-TEX significantly prolonged the survival time, the efficacy of DC-TEX exceeding that of the cytotoxic drugs. However, ATRA, Sun and most efficiently GEM, sufficed for a pronounced reduction of MDSC including tumor-infiltrating MDSC, which was accompanied by a decrease in migrating and metastasizing tumor cells. When combined with DC-TEX vaccination, a higher number of activated T cells was recovered in the tumor and the survival time was prolonged compared with only DC-TEX vaccinated mice. As ATRA, GEM and Sun affect MDSC at distinct maturation and activation stages, a stronger support for DC-TEX vaccination was expected by the drug combination. Intrapancreatic tumor growth was prevented beyond the death of control mice. However, tumors developed after a partial breakdown of the immune system by the persisting drug application. Nonetheless, in combination with optimized drug tuning to prevent MDSC maturation and activation, vaccination with TEX-loaded DC appears a most promising option in PaCa therapy.
Collapse
Affiliation(s)
- Li Xiao
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Ulrike Erb
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Kun Zhao
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Thilo Hackert
- Section Pancreas Research, University Hospital of Surgery, Heidelberg, Germany
| | - Margot Zöller
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| |
Collapse
|
38
|
Vienot A, Beinse G, Louvet C, de Mestier L, Meurisse A, Fein F, Heyd B, Cleau D, d’Engremont C, Dupont-Gossart AC, Lakkis Z, Tournigand C, Bouché O, Rousseau B, Neuzillet C, Bonnetain F, Borg C, Vernerey D. Overall Survival Prediction and Usefulness of Second-Line Chemotherapy in Advanced Pancreatic Adenocarcinoma. J Natl Cancer Inst 2017; 109:3105956. [DOI: 10.1093/jnci/djx037] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/21/2017] [Indexed: 02/07/2023] Open
|
39
|
Ebiike H, Taka N, Matsushita M, Ohmori M, Takami K, Hyohdoh I, Kohchi M, Hayase T, Nishii H, Morikami K, Nakanishi Y, Akiyama N, Shindoh H, Ishii N, Isobe T, Matsuoka H. Discovery of [5-Amino-1-(2-methyl-3H-benzimidazol-5-yl)pyrazol-4-yl]-(1H-indol-2-yl)methanone (CH5183284/Debio 1347), An Orally Available and Selective Fibroblast Growth Factor Receptor (FGFR) Inhibitor. J Med Chem 2016; 59:10586-10600. [DOI: 10.1021/acs.jmedchem.6b01156] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hirosato Ebiike
- Research
Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Naoki Taka
- Research
Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Masayuki Matsushita
- Research
Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Masayuki Ohmori
- Research
Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Kyoko Takami
- Research
Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Ikumi Hyohdoh
- Research
Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Masami Kohchi
- Research
Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Tadakatsu Hayase
- Research
Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Hiroki Nishii
- Research
Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Kenji Morikami
- Research
Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Yoshito Nakanishi
- Research
Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Nukinori Akiyama
- Research
Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Hidetoshi Shindoh
- Research
Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Nobuya Ishii
- Research
Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | - Takehito Isobe
- Research
Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Hiroharu Matsuoka
- Research
Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| |
Collapse
|
40
|
Vogel A, Ciardiello F, Hubner RA, Blanc JF, Carrato A, Yang Y, Patel DA, Ektare V, de Jong FA, Gill S. Post-gemcitabine therapy for patients with advanced pancreatic cancer - A comparative review of randomized trials evaluating oxaliplatin- and/or irinotecan-containing regimens. Cancer Treat Rev 2016; 50:142-147. [PMID: 27676174 DOI: 10.1016/j.ctrv.2016.09.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 09/01/2016] [Indexed: 02/07/2023]
Abstract
A systematic review and critical evaluation of randomized trial evidence for oxaliplatin- or irinotecan-containing regimens in patients with advanced pancreatic cancer previously treated with gemcitabine has not yet been published. We conducted a comparative systematic review of randomized trials evaluating oxaliplatin- or irinotecan-based therapies in patients with advanced pancreatic cancer previously treated with gemcitabine to assess trial similarity and the feasibility of performing an indirect treatment comparison (ITC). Studies were identified through PubMed and key oncology conference abstracts. The following trials met our criteria: NAPOLI-1 (nanoliposomal irinotecan [nal-IRI] or nal-IRI+5-fluorouracil [5-FU]/leucovorin [LV] vs 5-FU/LV), CONKO-003 (oxaliplatin+5-FU/LV [OFF] vs 5-FU/LV), PANCREOX (oxaliplatin+5-FU/LV [mFOLFOX6] vs 5-FU/LV), and Yoo et al. (2009) (irinotecan+5-FU/LV [mFOLFIRI3] vs mFOLFOX). Fundamental differences were identified in study design (i.e., number of study sites, number of countries), patient (i.e., locally advanced vs metastatic disease, stratification variables, prior and subsequent treatments) and treatment (i.e., regimens, dose intensity) characteristics, and primary and secondary outcomes (i.e., primary vs secondary outcomes, overall survival [OS], progression-free survival [PFS]) among the 4 included trials. Our comparative review demonstrated significant dissimilarity across trials, which precluded conducting an ITC. In the absence of head-to-head nal-IRI- and/or oxaliplatin-based therapy trials, clinicians are advised to interpret these studies separately within the context of their individual patient population.
Collapse
Affiliation(s)
- Arndt Vogel
- Medical School Hannover, Department of Gastroenterology, Hematology and Endocrinology, Carl-Neubergstr. 1, 30659 Hannover, Germany.
| | - Fortunato Ciardiello
- Dipartimento di Medicina Clinica e Sperimentale "F. Magrassi", Seconda Università degli Studi di Napoli, Via S. Pansini 5, 80131 Napoli, Italy.
| | - Richard A Hubner
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, United Kingdom.
| | - Jean-Frédéric Blanc
- Department of Hepato-Gastroenterology and Digestive Oncology, CHU Bordeaux, Hôpital Haut-Lévêque, avenue de Magellan, 33600 Pessac, France.
| | - Alfredo Carrato
- Medical Oncology Department, Ramon y Cajal University Hospital, IRYCIS, Alcala University, Carretera Colmenar Viejo km 9.1, 28034 Madrid, Spain.
| | - Yoojung Yang
- Shire, 650 East Kendall Street, Cambridge, MA 02145, United States.
| | - Dipen A Patel
- Pharmerit International, 4350 East West Highway, Suite 430, Bethesda, MD 20814, United States.
| | - Varun Ektare
- Pharmerit International, 4350 East West Highway, Suite 430, Bethesda, MD 20814, United States.
| | - Floris A de Jong
- Shire, Thurgauerstrasse 130, 8152 Glattpark (Opfikon), Zürich, Switzerland.
| | - Sharlene Gill
- University of British Columbia, BC Cancer Agency, 600 W. 10th Avenue, Vancouver, BC V5Z 4E6, Canada.
| |
Collapse
|
41
|
De Vita F, Ventriglia J, Febbraro A, Laterza MM, Fabozzi A, Savastano B, Petrillo A, Diana A, Giordano G, Troiani T, Conzo G, Galizia G, Ciardiello F, Orditura M. NAB-paclitaxel and gemcitabine in metastatic pancreatic ductal adenocarcinoma (PDAC): from clinical trials to clinical practice. BMC Cancer 2016; 16:709. [PMID: 27590845 PMCID: PMC5010686 DOI: 10.1186/s12885-016-2671-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/04/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma is an aggressive disease with poor prognosis. In a randomized phase III trial, combination of Nab-paclitaxel (Nab-P) plus gemcitabine showed superior activity and efficacy in first-line treatment compared with gemcitabine alone. METHODS Nab-P is not dispensed in Italy; however, we obtained this drug from our Ethics Committee for compassionate use. The aim of this study was to evaluate the efficacy and safety profile of this Nab-P and gemcitabine combination in a cohort of patients treated outside clinical trials. From January 2012 to May 2014, we included 41 patients with advanced pancreatic adenocarcinoma receiving combination of 125 mg/m(2) Nab-P and 1 g/m(2) gemcitabine on days 1, 8 and 15 of a 28-day cycle, as first-line treatment. Median age of patients was 67 (range 41-77) years, and 11 patients were aged ≥70 years. RESULTS Eastern Co-operative Oncology Group performance status was 0 or 1 in 32 patients (78 %) and 2 in nine patients (22 %). Primary tumor was located in the pancreatic head or body/tail in 24 (58.5 %) and 17 (41.5 %) patients, respectively, and nine patients had received biliary stent implantation before starting chemotherapy. Median carbohydrate antigen 19-9 level was 469 U/l (range 17.4-61546 U/l) and 29 patients (70.7 %) had referred pain at the time of diagnosis. Patients received a median six cycles (range 1-14) of treatment. Overall response rate was 36.6 %; median progression-free survival was 6.7 months [(95 % confidence interval (CI) 5.966-8.034), and median overall survival was 10 months (95 % CI 7.864-12.136). Treatment was well tolerated. No grade 4 toxicity was reported. Grade 3 toxicity included neutropenia in 10 patients (24.3 %), thrombocytopenia in five (12 %), anemia in three (7.3 %), diarrhea in four (9.7 %), nausea and vomiting in two (4.9 %), and fatigue in six (14.6 %). Finally, pain control was achieved in 24 of 29 patients (82.3 %) with a performance status improvement of 10 % according to the Karnofsky scale. CONCLUSIONS Our results confirm that combination of gemcitabine plus Nab-P is effective both in terms of overall response rate, progression-free survival and overall survival, with a good safety profile.
Collapse
Affiliation(s)
- Ferdinando De Vita
- Division of Medical Oncology, Department of Internal and Experimental Medicine “F. Magrassi”, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| | - Jole Ventriglia
- Division of Medical Oncology, Department of Internal and Experimental Medicine “F. Magrassi”, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| | - Antonio Febbraro
- Division of Medical Oncology, Fatebenefratelli Hospital, Viale Principe di Napoli 14/a, 82100 Benevento, Italy
| | - Maria Maddalena Laterza
- Division of Medical Oncology, Department of Internal and Experimental Medicine “F. Magrassi”, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| | - Alessio Fabozzi
- Division of Medical Oncology, Department of Internal and Experimental Medicine “F. Magrassi”, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| | - Beatrice Savastano
- Division of Medical Oncology, Department of Internal and Experimental Medicine “F. Magrassi”, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| | - Angelica Petrillo
- Division of Medical Oncology, Department of Internal and Experimental Medicine “F. Magrassi”, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| | - Anna Diana
- Division of Medical Oncology, Department of Internal and Experimental Medicine “F. Magrassi”, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| | - Guido Giordano
- Division of Medical Oncology, Fatebenefratelli Hospital, Viale Principe di Napoli 14/a, 82100 Benevento, Italy
| | - Teresa Troiani
- Division of Medical Oncology, Department of Internal and Experimental Medicine “F. Magrassi”, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| | - Giovanni Conzo
- Divisions of Surgical Oncology, Department of Anesthesiological, Surgical and Emergency Sciences, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| | - Gennaro Galizia
- Divisions of Surgical Oncology, Department of Anesthesiological, Surgical and Emergency Sciences, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| | - Fortunato Ciardiello
- Division of Medical Oncology, Department of Internal and Experimental Medicine “F. Magrassi”, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| | - Michele Orditura
- Division of Medical Oncology, Department of Internal and Experimental Medicine “F. Magrassi”, Second University of Naples - School of Medicine, c/o II Policlinico, Via Pansini, 5, 80131 Naples, Italy
| |
Collapse
|
42
|
Bupathi M, Ahn DH, Wu C, Ciombor KK, Stephens JA, Reardon J, Goldstein DA, Bekaii-Saab T. Modified irinotecan and infusional 5-fluorouracil (mFOLFIRI) in patients with refractory advanced pancreas cancer (APC): a single-institution experience. Med Oncol 2016; 33:37. [PMID: 26995224 PMCID: PMC4976592 DOI: 10.1007/s12032-016-0753-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/04/2016] [Indexed: 12/18/2022]
Abstract
Pancreatic adenocarcinoma is the fourth leading cause of cancer death. Recently, MM-398 (nanoliposomal irinotecan) was shown to be associated with significant improvement in outcome measures with acceptable toxicities when combined with 5-fluorouracil (5-FU)/leucovorin (LV) compared to 5-FU/LV alone in patients failing one line of gemcitabine-based therapy. There is a paucity of data evaluating the role of irinotecan in combination with 5FU in advanced pancreas cancer (APC). We performed a retrospective analysis of all patients who received mFOLFIRI (minus bolus 5FU and LV). All patients with metastatic disease who had failed at least one line of gemcitabine-based therapy prior to receiving mFOLFIRI were included in this study. Descriptive statistics were used to assess the continuous variables and adverse events (AEs), and Kaplan-Meier methods were used to calculate the median progression-free survival (PFS) and overall survival (OS). Forty patients were included in this analysis. Patients received 1-5 lines of prior therapy (25 % with more than 3 lines of prior therapy). The mean age at diagnosis was 60, and 98 % had ECOG of 1. The mean CA 19-9 at the start of therapy was 33,169 U/ml. The median PFS was 2.59 months [95 % confidence interval (CI) (1.90, 3.54)], and OS was 4.75 months [95 % CI (3.14, 8.98)]. The most common AEs included fatigue (98 %), neuropathy (83 %), anorexia (68 %), nausea (60 %) and constipation (55 %). Grade 3 toxicities included fatigue (13 %) and rash (3 %). There were no observed grade 4 toxicities. In this single-institution retrospective analysis, mFOLFIRI was found to be both tolerable and relatively effective in a heavily pretreated patient population with APC. Future prospective studies should consider evaluating the role of mFOLFIRI in refractory APC.
Collapse
Affiliation(s)
- M Bupathi
- Department of Medical Oncology, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University Wexner Medical Center, A454 Startling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43210, USA
| | - D H Ahn
- Department of Medical Oncology, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University Wexner Medical Center, A454 Startling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43210, USA
| | - C Wu
- Department of Medical Oncology, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University Wexner Medical Center, A454 Startling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43210, USA
| | - K K Ciombor
- Department of Medical Oncology, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University Wexner Medical Center, A454 Startling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43210, USA
| | - J A Stephens
- Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - J Reardon
- Department of Pharmacy, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University, Columbus, OH, USA
| | - D A Goldstein
- Winship Cancer Institute, Emory University, 1365-C Clifton Rd NE, Atlanta, GA, USA
- Davidoff Cancer Center, Rabin Medical Center, Derech Ze'ev Jabotinsky 39, 4941492, Petach Tikva, Israel
| | - T Bekaii-Saab
- Department of Medical Oncology, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University Wexner Medical Center, A454 Startling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
43
|
Wang-Gillam A, Li CP, Bodoky G, Dean A, Shan YS, Jameson G, Macarulla T, Lee KH, Cunningham D, Blanc JF, Hubner RA, Chiu CF, Schwartsmann G, Siveke JT, Braiteh F, Moyo V, Belanger B, Dhindsa N, Bayever E, Von Hoff DD, Chen LT. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet 2016; 387:545-557. [PMID: 26615328 DOI: 10.1016/s0140-6736(15)00986-1] [Citation(s) in RCA: 766] [Impact Index Per Article: 85.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Nanoliposomal irinotecan showed activity in a phase 2 study in patients with metastatic pancreatic ductal adenocarcinoma previously treated with gemcitabine-based therapies. We assessed the effect of nanoliposomal irinotecan alone or combined with fluorouracil and folinic acid in a phase 3 trial in this population. METHODS We did a global, phase 3, randomised, open-label trial at 76 sites in 14 countries. Eligible patients with metastatic pancreatic ductal adenocarcinoma previously treated with gemcitabine-based therapy were randomly assigned (1:1) using an interactive web response system at a central location to receive either nanoliposomal irinotecan monotherapy (120 mg/m(2) every 3 weeks, equivalent to 100 mg/m(2) of irinotecan base) or fluorouracil and folinic acid. A third arm consisting of nanoliposomal irinotecan (80 mg/m(2), equivalent to 70 mg/m(2) of irinotecan base) with fluorouracil and folinic acid every 2 weeks was added later (1:1:1), in a protocol amendment. Randomisation was stratified by baseline albumin, Karnofsky performance status, and ethnic origin. Treatment was continued until disease progression or intolerable toxic effects. The primary endpoint was overall survival, assessed in the intention-to-treat population. The primary analysis was planned after 305 events. Safety was assessed in all patients who had received study drug. This trial is registered at ClinicalTrials.gov, number NCT01494506. FINDINGS Between Jan 11, 2012, and Sept 11, 2013, 417 patients were randomly assigned either nanoliposomal irinotecan plus fluorouracil and folinic acid (n=117), nanoliposomal irinotecan monotherapy (n=151), or fluorouracil and folinic acid (n=149). After 313 events, median overall survival in patients assigned nanoliposomal irinotecan plus fluorouracil and folinic acid was 6.1 months (95% CI 4.8-8.9) vs 4.2 months (3.3-5.3) with fluorouracil and folinic acid (hazard ratio 0.67, 95% CI 0.49-0.92; p=0.012). Median overall survival did not differ between patients assigned nanoliposomal irinotecan monotherapy and those allocated fluorouracil and folinic acid (4.9 months [4.2-5.6] vs 4.2 months [3.6-4.9]; 0.99, 0.77-1.28; p=0.94). The grade 3 or 4 adverse events that occurred most frequently in the 117 patients assigned nanoliposomal irinotecan plus fluorouracil and folinic acid were neutropenia (32 [27%]), diarrhoea (15 [13%]), vomiting (13 [11%]), and fatigue (16 [14%]). INTERPRETATION Nanoliposomal irinotecan in combination with fluorouracil and folinic acid extends survival with a manageable safety profile in patients with metastatic pancreatic ductal adenocarcinoma who previously received gemcitabine-based therapy. This agent represents a new treatment option for this population. FUNDING Merrimack Pharmaceuticals.
Collapse
Affiliation(s)
| | - Chung-Pin Li
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital and National Yang-Ming University School of Medicine, Taipei, Taiwan
| | | | - Andrew Dean
- St John of God Hospital, Subiaco, WA, Australia
| | - Yan-Shen Shan
- National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | | | - Teresa Macarulla
- Vall d'Hebron University Hospital (HUVH) and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Kyung-Hun Lee
- Seoul National University Hospital, Seoul, South Korea
| | | | | | | | | | | | - Jens T Siveke
- Klinikum rechts der Isar der T U München, Munich, Germany
| | - Fadi Braiteh
- Comprehensive Cancer Centers of Nevada, Las Vegas, NV, USA
| | - Victor Moyo
- Merrimack Pharmaceuticals, Cambridge, MA, USA
| | | | | | | | | | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, and Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
44
|
Ko AH, Bekaii-Saab T, Van Ziffle J, Mirzoeva OM, Joseph NM, Talasaz A, Kuhn P, Tempero MA, Collisson EA, Kelley RK, Venook AP, Dito E, Ong A, Ziyeh S, Courtin R, Linetskaya R, Tahiri S, Korn WM. A Multicenter, Open-Label Phase II Clinical Trial of Combined MEK plus EGFR Inhibition for Chemotherapy-Refractory Advanced Pancreatic Adenocarcinoma. Clin Cancer Res 2016; 22:61-8. [PMID: 26251290 PMCID: PMC4703532 DOI: 10.1158/1078-0432.ccr-15-0979] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/28/2015] [Indexed: 12/21/2022]
Abstract
PURPOSE On the basis of preclinical evidence of synergistic activity between MEK and EGFR inhibitors in pancreatic ductal adenocarcinoma (PDAC), we evaluated the safety and efficacy of selumetinib, a MEK1/2 inhibitor, plus erlotinib in patients with previously treated advanced PDAC. EXPERIMENTAL DESIGN In this single-arm phase II trial, eligible patients received the combination of erlotinib 100 mg plus selumetinib 100 mg daily in 3-week cycles. Study assessments included measurement of clinical outcomes, with a primary endpoint of overall survival, and exploration of potential molecular predictors of treatment benefit. RESULTS Forty-six patients were enrolled and received a median of two cycles (range, 1-7). Although no objective responses were observed, 19 patients (41%) showed evidence of stable disease for ≥6 weeks, and 13 of 34 patients (38%) had a CA19-9 decline ≥50%. Median progression-free survival was 1.9 months [95% confidence interval (CI), 1.4-3.3 months], with a median overall survival of 7.3 months (95% CI, 5.2-8.0 months). Common adverse events included rash, diarrhea, and nausea/vomiting. Patients with tumors exhibiting an epithelial phenotype (demonstrated by a high level of E-cadherin expression) were more likely to be sensitive to study treatment. Tumor-derived DNA was detectable in plasma from the majority of patients using next-generation digital DNA sequencing, and its relative abundance correlated with tumor burden. CONCLUSIONS A therapeutic strategy of dual targeted inhibition of the MEK and EGFR pathways shows modest antitumor activity in pancreatic cancer. Specific molecular subtypes may derive greatest benefit from this combination. Further exploration, both with more potent MEK inhibitors and in molecularly enriched patient subsets, is warranted.
Collapse
Affiliation(s)
- Andrew H Ko
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California.
| | | | - Jessica Van Ziffle
- Department of Pathology, University of California San Francisco, San Francisco, California
| | - Olga M Mirzoeva
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Nancy M Joseph
- Department of Pathology, University of California San Francisco, San Francisco, California
| | | | - Peter Kuhn
- University of Southern California, Los Angeles, California
| | - Margaret A Tempero
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Eric A Collisson
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - R Kate Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Alan P Venook
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Elizabeth Dito
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Anna Ong
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Sharvina Ziyeh
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Ryan Courtin
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Regina Linetskaya
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Sanaa Tahiri
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - W Michael Korn
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| |
Collapse
|
45
|
Curaxin CBL0137 eradicates drug resistant cancer stem cells and potentiates efficacy of gemcitabine in preclinical models of pancreatic cancer. Oncotarget 2015; 5:11038-53. [PMID: 25402820 PMCID: PMC4294371 DOI: 10.18632/oncotarget.2701] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 11/06/2014] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) continues to be one of the deadliest cancers due to the absence of effective treatment. Curaxins are a class of small molecules with anti-cancer activity demonstrated in different models of cancer in mice. The lead curaxin compound, CBL0137, recently entered Phase I clinical trials. Curaxins modulate several important signaling pathways involved in the pathogenesis of PDA through inhibition of chromatin remodeling complex FACT. FACT is overexpressed in multiple types of tumor, with one of the highest rate of overexpression in PDA (59%). In this study, the efficacy of CBL0137 alone or in combination with current standard of care, gemcitabine, was tested against different models of PDA in vitro and in mouse models. It was found that CBL0137 alone is a potent inducer of apoptosis in pancreatic cancer cell lines and is toxic not only for proliferating bulk tumor cells, but also for pancreatic cancer stem cells. In mice, CBL0137 was effective against several PDA models, including orthotopic gemcitabine resistant PANC-1 model and patient derived xenografts, in which CBL0137 anti-tumor effect correlated with overexpression of FACT. Moreover, we observed synergy of CBL0137 with gemcitabine which may be explained by the ability of CBL0137 to inhibit several transcriptional programs induced by gemcitabine, including NF-kappaB response and expression of ribonucleotide reductase, one of the targets of gemcitabine in cells. This data suggest testing of CBL0137 efficacy in Phase II trial in PDA patients alone and in combination with gemcitabine.
Collapse
|
46
|
Neuzillet C, Tijeras-Raballand A, Bourget P, Cros J, Couvelard A, Sauvanet A, Vullierme MP, Tournigand C, Hammel P. State of the art and future directions of pancreatic ductal adenocarcinoma therapy. Pharmacol Ther 2015; 155:80-104. [PMID: 26299994 DOI: 10.1016/j.pharmthera.2015.08.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/17/2015] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is expected to become the second cause of cancer-related death in 2030. PDAC is the poorest prognostic tumor of the digestive tract, with 80% of patients having advanced disease at diagnosis and 5-year survival rate not exceeding 7%. Until 2010, gemcitabine was the only validated therapy for advanced PDAC with a modest improvement in median overall survival as compared to best supportive care (5-6 vs 3 months). Multiple phase II-III studies have used various combinations of gemcitabine with other cytotoxics or targeted agents, most in vain, in attempt to improve this outcome. Over the past few years, the landscape of PDAC management has undergone major and rapid changes with the approval of the FOLFIRINOX and gemcitabine plus nab-paclitaxel regimens in patients with metastatic disease. These two active combination chemotherapy options yield an improved median overall survival (11.1 vs 8.5 months, respectively) thus making longer survival a reasonably achievable goal. This breakthrough raises some new clinical questions about the management of PDAC. Moreover, better knowledge of the environmental and genetic events that underpin multistep carcinogenesis and of the microenvironment surrounding cancer cells in PDAC has open new perspectives and therapeutic opportunities. In this new dynamic context of deep transformation in basic research and clinical management aspects of the disease, we gathered updated preclinical and clinical data in a multifaceted review encompassing the lessons learned from the past, the yet unanswered questions, and the most promising research priorities to be addressed for the next 5 years.
Collapse
Affiliation(s)
- Cindy Neuzillet
- INSERM UMR1149, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Digestive Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Medical Oncology, Henri Mondor University Hospital, 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France.
| | - Annemilaï Tijeras-Raballand
- Department of Translational Research, AAREC Filia Research, 1 place Paul Verlaine, 92100 Boulogne-Billancourt, France
| | - Philippe Bourget
- Department of Clinical Pharmacy, Necker-Enfants Malades University Hospital, 149 Rue de Sèvres, 75015 Paris, France
| | - Jérôme Cros
- INSERM UMR1149, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Pathology, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France
| | - Anne Couvelard
- INSERM UMR1149, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Pathology, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France
| | - Alain Sauvanet
- Department of Biliary and Pancreatic Surgery, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, 92110 Clichy, France
| | - Marie-Pierre Vullierme
- Department of Radiology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, 92110 Clichy, France
| | - Christophe Tournigand
- Department of Medical Oncology, Henri Mondor University Hospital, 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Pascal Hammel
- INSERM UMR1149, Bichat-Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 46 rue Henri Huchard, 75018 Paris, and 100 boulevard du Général Leclerc, 92110 Clichy, France; Department of Digestive Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, 92110 Clichy, France
| |
Collapse
|
47
|
Gall TMH, Tsakok M, Wasan H, Jiao LR. Pancreatic cancer: current management and treatment strategies. Postgrad Med J 2015; 91:601-7. [PMID: 26243882 DOI: 10.1136/postgradmedj-2014-133222] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 07/14/2015] [Indexed: 12/18/2022]
Abstract
The 5-year survival of patients with pancreatic cancer is poor and, despite oncological advances over the past two decades, has not significantly improved. However, there have been several surgical and oncological advances which have improved morbidity and mortality in surgery and more efficacious chemotherapy regimens, resulting in a better patient experience and an increase in survival by a number of months. Most patients have a tumour at the head of the pancreas and those with resectable disease undergo a pancreaticoduodenectomy, which can be performed laparoscopically. Those who have a pancreatic resection have an increased survival in comparison with those receiving oncological treatment only; however, only a quarter of patients have resectable disease at diagnosis. Some centres are now performing venous resections and/or arterial resections in order to increase the number of patients eligible for curative surgery. Innovative techniques using ablation technologies to downstage tumours for resection are also being investigated. After surgery, all patients should be offered adjuvant gemcitabine-based chemotherapy. Those with locally advanced tumours not suitable for surgery should be offered FOLFIRINOX chemotherapy, after which the tumour may be suitable for surgical resection. The use of radiotherapy in this group of patients is controversial but offered by a few centres. Patients with metastatic disease at diagnosis should also be offered FOLFIRINOX chemotherapy, which can improve survival by a few months. As our knowledge of the tumour biology of pancreatic cancer progresses, a number of new agents targeting specific genes and proteins are under investigation and there is hope that median survival will continue to improve over the next decade.
Collapse
Affiliation(s)
- Tamara M H Gall
- HPB Surgical Unit, Department of Surgery & Cancer, Imperial College, Hammersmith Hospital Campus, London, UK
| | - Maria Tsakok
- HPB Surgical Unit, Department of Surgery & Cancer, Imperial College, Hammersmith Hospital Campus, London, UK
| | - Harpreet Wasan
- Department of Oncology, Imperial College, Hammersmith Hospital Campus, London, UK
| | - Long R Jiao
- HPB Surgical Unit, Department of Surgery & Cancer, Imperial College, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
48
|
Tröger W, Galun D, Reif M, Schumann A, Stanković N, Milićević M. Quality of life of patients with advanced pancreatic cancer during treatment with mistletoe: a randomized controlled trial. DEUTSCHES ARZTEBLATT INTERNATIONAL 2015; 111:493-502, 33 p following 502. [PMID: 25142075 DOI: 10.3238/arztebl.2014.0493] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 12/02/2013] [Accepted: 05/09/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND The treatment of cancer patients with mistletoe extract is said to prolong their survival and, above all, improve their quality of life. We studied whether the quality of life of patients with advanced pancreatic cancer could be improved by mistletoe extract. METHOD An open, single-center, group-sequential, randomized phase III trial (ISRCTN70760582) was conducted. From January 2009 to December 2010, 220 patients with locally advanced or metastatic pancreatic cancer who were receiving no further treatment for pancreatic cancer other than best supportive care were included in this trial. They were stratified by prognosis and randomly allocated either to a group that received mistletoe treatment or to one that did not. Mistletoe extract was given in escalating doses by subcutaneous injection three times a week. The planned interim evaluation of data from 220 patients indicated that mistletoe treatment was associated with longer overall survival, and the trial was terminated prematurely. After termination of the study, the results with respect to quality of life (assessed with the QLO-C30 scales of the European Organisation for Research and Treatment of Cancer) and trends in body weight were evaluated. RESULTS Data on quality of life and body weight were obtained from 96 patients treated with mistletoe and 72 control patients. Those treated with mistletoe did better on all 6 functional scales and on 7 of 9 symptom scales, including pain (95% confidence interval [CI] -29 to -17), fatigue (95% CI -36.1 to -25.0), appetite loss (95% CI -51 to -36.7), and insomnia (95% CI -45.8 to -28.6). This is reflected by the trend in body weight during the trial. CONCLUSION In patients with locally advanced or metastatic pancreatic carcinoma, mistletoe treatment significantly improves the quality of life in comparison to best supportive care alone. Mistletoe is an effective second-line treatment for this disease.
Collapse
Affiliation(s)
| | - Danijel Galun
- First Surgical Clinic of the Clinical Centers of Servia (Belgrade)
| | | | | | | | | |
Collapse
|
49
|
Qin S, Deng Y, Li J, Zhang Z. A bioengineered murine model using CD24
+
CD44
+
pancreatic cancer stem cells for chemotherapy study. Biomed Mater 2014; 10:015004. [DOI: 10.1088/1748-6041/10/1/015004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
50
|
Pan Y, Rhea P, Tan L, Cartwright C, Lee HJ, Ravoori MK, Addington C, Gagea M, Kundra V, Kim SJ, Newman RA, Yang P. PBI-05204, a supercritical CO₂ extract of Nerium oleander, inhibits growth of human pancreatic cancer via targeting the PI3K/mTOR pathway. Invest New Drugs 2014; 33:271-9. [PMID: 25476893 PMCID: PMC4387257 DOI: 10.1007/s10637-014-0190-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 11/10/2014] [Indexed: 01/03/2023]
Abstract
Introduction Oleandrin, a cardiac glycoside, exerts strong anti-proliferative activity against various human malignancies in in vitro cells. Here, we report the antitumor efficacy of PBI-05204, a supercritical C0₂ extract of Nerium oleander containing oleandrin, in a human pancreatic cancer Panc-1 orthotopic model. Results While all the control mice exhibited tumors by the end of treatment, only 2 of 8 mice (25%) treated for 6 weeks with PBI-05204 (40 mg/kg) showed dissectible tumor at the end of the treatment period. The average tumor weight (222.9 ± 116.9 mg) in mice treated with PBI-05204 (20 mg/kg) was significantly reduced from that in controls (920.0 ± 430.0 mg) (p < 0.05). Histopathologic examination of serial sections from each pancreas with no dissectible tumor in the PBI-05204 (40 mg/kg) treated group showed that the pancreatic tissues of 5/6 mice were normal while the remaining mouse had a tumor the largest diameter of which was less than 2.3 mm. In contrast, while gemcitabine alone did not significantly reduce tumor growth, PBI-05204 markedly enhanced the antitumor efficacy of gemcitabine in this particular model. Ki-67 staining was reduced in pancreatic tumors from mice treated with PBI-05204 (20 mg/kg) compared to that of control, suggesting that PBI-05204 inhibited the proliferation of the Panc-1 tumor cells. PBI-05204 suppressed expression of pAkt, pS6, and p4EPB1 in a concentration-dependent manner in both Panc-1 tumor tissues and human pancreatic cancer cell lines, implying that this novel botanical drug exerts its potent antitumor activity, at least in part, through down-regulation of PI3k/Akt and mTOR pathways.
Collapse
Affiliation(s)
- Yong Pan
- Department of General Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 0462, Houston, TX, 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|