1
|
Haggstrom L, Chan WY, Nagrial A, Chantrill LA, Sim HW, Yip D, Chin V. Chemotherapy and radiotherapy for advanced pancreatic cancer. Cochrane Database Syst Rev 2024; 12:CD011044. [PMID: 39635901 PMCID: PMC11619003 DOI: 10.1002/14651858.cd011044.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is a lethal disease with few effective treatment options. Many anti-cancer therapies have been tested in the locally advanced and metastatic setting, with mixed results. This review synthesises all the randomised data available to help better inform patient and clinician decision-making. It updates the previous version of the review, published in 2018. OBJECTIVES To assess the effects of chemotherapy, radiotherapy, or both on overall survival, severe or life-threatening adverse events, and quality of life in people undergoing first-line treatment of advanced pancreatic cancer. SEARCH METHODS We searched for published and unpublished studies in CENTRAL, MEDLINE, Embase, and CANCERLIT, and handsearched various sources for additional studies. The latest search dates were in March and July 2023. SELECTION CRITERIA We included randomised controlled trials comparing chemotherapy, radiotherapy, or both with another intervention or best supportive care. Participants were required to have locally advanced, unresectable pancreatic cancer or metastatic pancreatic cancer not amenable to curative intent treatment. Histological confirmation was required. Trials were required to report overall survival. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included 75 studies in the review and 51 in the meta-analysis (11,333 participants). We divided the studies into seven categories: any anti-cancer treatment versus best supportive care; various chemotherapy types versus gemcitabine; gemcitabine-based combinations versus gemcitabine alone; various chemotherapy combinations versus gemcitabine plus nab-paclitaxel; fluoropyrimidine-based studies; miscellaneous studies; and radiotherapy studies. In general, the included studies were at low risk for random sequence generation, detection bias, attrition bias, and reporting bias, at unclear risk for allocation concealment, and high risk for performance bias. Compared to best supportive care, chemotherapy likely results in little to no difference in overall survival (OS) (hazard ratio (HR) 1.08, 95% confidence interval (CI) 0.88 to 1.33; absolute risk of death at 12 months of 971 per 1000 versus 962 per 1000; 4 studies, 298 participants; moderate-certainty evidence). The adverse effects of chemotherapy and impacts on quality of life (QoL) were uncertain. Many of the chemotherapy regimens were outdated. Eight studies compared non-gemcitabine-based chemotherapy regimens to gemcitabine. These showed that 5-fluorouracil (5FU) likely reduces OS (HR 1.69, 95% CI 1.26 to 2.27; risk of death at 12 months of 914 per 1000 versus 767 per 1000; 1 study, 126 participants; moderate certainty), and grade 3/4 adverse events (QoL not reported). Fixed dose rate gemcitabine likely improves OS (HR 0.79, 95% CI 0.66 to 0.94; risk of death at 12 months of 683 per 1000 versus 767 per 1000; 2 studies, 644 participants; moderate certainty), and likely increase grade 3/4 adverse events (QoL not reported). FOLFIRINOX improves OS (HR 0.51, 95% CI 0.43 to 0.60; risk of death at 12 months of 524 per 1000 versus 767 per 1000; P < 0.001; 2 studies, 652 participants; high certainty), and delays deterioration in QoL, but increases grade 3/4 adverse events. Twenty-eight studies compared gemcitabine-based combinations to gemcitabine. Gemcitabine plus platinum may result in little to no difference in OS (HR 0.94, 95% CI 0.81 to 1.08; risk of death at 12 months of 745 per 1000 versus 767 per 1000; 6 studies, 1140 participants; low certainty), may increase grade 3/4 adverse events, and likely worsens QoL. Gemcitabine plus fluoropyrimidine improves OS (HR 0.88, 95% CI 0.81 to 0.95; risk of death at 12 months of 722 per 1000 versus 767 per 1000; 10 studies, 2718 participants; high certainty), likely increases grade 3/4 adverse events, and likely improves QoL. Gemcitabine plus topoisomerase inhibitors result in little to no difference in OS (HR 1.01, 95% CI 0.87 to 1.16; risk of death at 12 months of 770 per 1000 versus 767 per 1000; 3 studies, 839 participants; high certainty), likely increases grade 3/4 adverse events, and likely does not alter QoL. Gemcitabine plus taxane result in a large improvement in OS (HR 0.71, 95% CI 0.62 to 0.81; risk of death at 12 months of 644 per 1000 versus 767 per 1000; 2 studies, 986 participants; high certainty), and likely increases grade 3/4 adverse events and improves QoL. Nine studies compared chemotherapy combinations to gemcitabine plus nab-paclitaxel. Fluoropyrimidine-based combination regimens improve OS (HR 0.79, 95% CI 0.70 to 0.89; risk of death at 12 months of 542 per 1000 versus 628 per 1000; 6 studies, 1285 participants; high certainty). The treatment arms had distinct toxicity profiles, and there was little to no difference in QoL. Alternative schedules of gemcitabine plus nab-paclitaxel likely result in little to no difference in OS (HR 1.10, 95% CI 0.82 to 1.47; risk of death at 12 months of 663 per 1000 versus 628 per 1000; 2 studies, 367 participants; moderate certainty) or QoL, but may increase grade 3/4 adverse events. Four studies compared fluoropyrimidine-based combinations to fluoropyrimidines alone, with poor quality evidence. Fluoropyrimidine-based combinations are likely to result in little to no impact on OS (HR 0.84, 95% CI 0.61 to 1.15; risk of death at 12 months of 765 per 1000 versus 704 per 1000; P = 0.27; 4 studies, 491 participants; moderate certainty) versus fluoropyrimidines alone. The evidence suggests that there was little to no difference in grade 3/4 adverse events or QoL between the two groups. We included only one radiotherapy (iodine-125 brachytherapy) study with 165 participants. The evidence is very uncertain about the effect of radiotherapy on outcomes. AUTHORS' CONCLUSIONS Combination chemotherapy remains standard of care for metastatic pancreatic cancer. Both FOLFIRINOX and gemcitabine plus a taxane improve OS compared to gemcitabine alone. Furthermore, the evidence suggests that fluoropyrimidine-based combination chemotherapy regimens improve OS compared to gemcitabine plus nab-paclitaxel. The effects of radiotherapy were uncertain as only one low-quality trial was included. Selection of the most appropriate chemotherapy for individuals still remains unpersonalised, with clinicopathological stratification remaining elusive. Biomarker development is essential to assist in rationalising treatment selection for patients.
Collapse
Affiliation(s)
- Lucy Haggstrom
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Wei Yen Chan
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Sydney, Australia
| | - Adnan Nagrial
- The Crown Princess Mary Cancer Centre, Westmead, Australia
- Medical School, The University of Sydney, Sydney, Australia
| | - Lorraine A Chantrill
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- University of Wollongong, Wollongong, Australia
| | - Hao-Wen Sim
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Desmond Yip
- Department of Medical Oncology, The Canberra Hospital, Garran, Australia
- ANU Medical School, Australian National University, Acton, Australia
| | - Venessa Chin
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- Medical Oncology, Garvan Institute of Medical Research, Sydney, Australia
| |
Collapse
|
2
|
Tushoski-Alemán GW, Crespin AJ, Oguejiofor CJ, Szymkiewicz DD, Herremans KM, Han S, Hughes SJ. Variability of quality-of-life measurements and reporting in randomised controlled trials of pancreatic cancer: a systematic review. BMJ Open 2024; 14:e083696. [PMID: 39551595 PMCID: PMC11574412 DOI: 10.1136/bmjopen-2023-083696] [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: 12/26/2023] [Accepted: 10/11/2024] [Indexed: 11/19/2024] Open
Abstract
OBJECTIVES This systematic review aims to evaluate the methodology used in pancreatic cancer (PC) randomised controlled trials (RCTs) measuring quality of life (QOL) and focuses on the type, frequency, survey compliance and duration of these assessments. DESIGN Systematic review of PC RCTs measuring QOL. DATA SOURCES A search of PubMed.gov and ClinicalTrials.gov was conducted for PC RCTs measuring QOL from inception to 21 March 2023. Only phase III RCTs were included. Studies were excluded if QOL was not measured, the study was phase I/II, in the second-line setting or unavailable in English. Data were independently extracted by two reviewers in a standardised fashion. PRIMARY AND SECONDARY OUTCOME MEASURES Primary outcomes included the type of QOL instrument used, the timing and frequency of assessments, methods of analysis and survey completion rates (SCRs) over time. Secondary outcomes included patient demographics, significant QOL improvements and the frequency of trials measuring QOL. RESULTS Out of 269 studies screened, 54 RCTs were identified, and 24 measured QOL (involving 11 229 patients). Instruments used included the EORTC QLQ-C30 (n=15), FACT-HEP (n=3), Spitzer-QOL-Index (n=2), EQ-5D (n=2), LASA (n=1) and FACT-PA (n=1). Most trials assessed QOL until disease progression or death (10/24), with 4-week intervals being the most common (7/24). SCRs were reported in 15/24 trials, with disease stage influencing SCRs over time. In trials with metastatic, locally advanced/metastatic, and resectable disease, the median times to reach a 50% response rate-defined as the point where the number of surveys completed was half of the enrolled participants-were 12.41 weeks (n=2), 14.14 weeks (n=10), and 54.2 weeks (n=3), respectively." Only 2/24 trials reported significant QOL improvements between treatment arms. Patient age was reported in all trials, while race/ethnicity was only reported in 4/24 trials. CONCLUSIONS Significant variability exists in the timing, methods and reporting of QOL assessments in PC trials. There is a need for further research to assess the implications of missing data and consider the temporality of QOL assessment in patients with advanced cancers and poor prognosis.
Collapse
Affiliation(s)
| | | | | | | | - Kelly M Herremans
- Department of Surgery, University of Florida, Gainesville, Florida, USA
| | - Song Han
- Department of Surgery, University of Florida, Gainesville, Florida, USA
| | - Steven J Hughes
- Department of Surgery, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
3
|
Wang J, Yang J, Narang A, He J, Wolfgang C, Li K, Zheng L. Consensus, debate, and prospective on pancreatic cancer treatments. J Hematol Oncol 2024; 17:92. [PMID: 39390609 PMCID: PMC11468220 DOI: 10.1186/s13045-024-01613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Pancreatic cancer remains one of the most aggressive solid tumors. As a systemic disease, despite the improvement of multi-modality treatment strategies, the prognosis of pancreatic cancer was not improved dramatically. For resectable or borderline resectable patients, the surgical strategy centered on improving R0 resection rate is consensus; however, the role of neoadjuvant therapy in resectable patients and the optimal neoadjuvant therapy of chemotherapy with or without radiotherapy in borderline resectable patients were debated. Postoperative adjuvant chemotherapy of gemcitabine/capecitabine or mFOLFIRINOX is recommended regardless of the margin status. Chemotherapy as the first-line treatment strategy for advanced or metastatic patients included FOLFIRINOX, gemcitabine/nab-paclitaxel, or NALIRIFOX regimens whereas 5-FU plus liposomal irinotecan was the only standard of care second-line therapy. Immunotherapy is an innovative therapy although anti-PD-1 antibody is currently the only agent approved by for MSI-H, dMMR, or TMB-high solid tumors, which represent a very small subset of pancreatic cancers. Combination strategies to increase the immunogenicity and to overcome the immunosuppressive tumor microenvironment may sensitize pancreatic cancer to immunotherapy. Targeted therapies represented by PARP and KRAS inhibitors are also under investigation, showing benefits in improving progression-free survival and objective response rate. This review discusses the current treatment modalities and highlights innovative therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Junke Wang
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jie Yang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Amol Narang
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jin He
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Christopher Wolfgang
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York, NY, USA
| | - Keyu Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Multidisciplinary Gastrointestinal Cancer Laboratories Program, the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
4
|
Abstract
Pancreatic cancer remains among the malignancies with the worst outcomes. Survival has been improving, but at a slower rate than other cancers. Multimodal treatment, including chemotherapy, surgical resection, and radiotherapy, has been under investigation for many years. Because of the anatomical characteristics of the pancreas, more emphasis on treatment selection has been placed on local extension into major vessels. Recently, the development of more effective treatment regimens has opened up new treatment strategies, but urgent research questions have also become apparent. This review outlines the current management of pancreatic cancer, and the recent advances in its treatment. The review discusses future treatment pathways aimed at integrating novel findings of translational and clinical research.
Collapse
Affiliation(s)
- Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Toshitaka Sugawara
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sana D Karam
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Wells A Messersmith
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
5
|
Hau SO, Svensson M, Petersson A, Eberhard J, Jirström K. Trajectories of immune-related serum proteins and quality of life in patients with pancreatic and other periampullary cancer: the CHAMP study. BMC Cancer 2023; 23:1074. [PMID: 37936126 PMCID: PMC10629201 DOI: 10.1186/s12885-023-11562-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND There is still a profound lack of efficient therapeutic strategies against pancreatic and other periampullary adenocarcinoma. Surgery is seldom possible, leaving palliative chemotherapy the only option for most patients. Chemotherapy treatment is however often accompanied by serious side-effects, and the identification of biomarkers for early prediction of disease and treatment-associated symptoms could help alleviate patient suffering. This study investigated the dynamic interrelationship between immune-related serum proteins, routine biomarkers, and health-related quality of life (HRQoL) factors during chemotherapy treatment of patients enrolled in the prospective, observational study Chemotherapy, Host response And Molecular dynamics in Periampullary cancer (CHAMP). METHODS Proximity extension assay was applied to analyse 92 immune-associated proteins in longitudinal serum samples from 75 patients, 18 treated with curative and 57 with palliative intent. HRQoL data were available from all patients at baseline (BL), from 41 patients at three months, and from 23 patients at six months. Information on routine laboratory parameters albumin, CA19-9, CEA and CRP were collected from medical charts. RESULTS In total nine proteins; chemokine (C-C motif) ligand 23 (CCL23), cluster of differentiation 4 (CD4), cluster of differentiation 28 (CD28), decorin (DCN), galectin-1 (Gal-1), granzyme B (GZMB), granzyme H (GZMH), matrix metallopeptidase 7 (MMP7), and monocyte chemotactic protein-1 (MCP-1) were strongly correlated (Spearman's Rho ≤ -0.6 or ≥ 0.6) with either cognitive functioning (DCN), emotional functioning (DCN, MCP-1), dyspnoea (CD28, GZMB, GZMH) or insomnia (CCL23, CD4, Gal-1, MMP7) during treatment. Associations between routine laboratory parameters (CA 19-9, CA-125, CRP, CEA and albumin) and HRQoL factors were overall weaker. None of the investigated proteins were associated with pain. CONCLUSIONS This is, to our knowledge, the first study exploring associations between serum biomarkers and HRQoL in patients with pancreatic or other periampullary cancer, and some findings merit further validation. The associations of DCN and MCP-1with impaired cognitive and/or emotional functioning are of particular interest, given their established link to various neurodegenerative conditions. Chemotherapy is known to cause persistent cognitive dysfunction with effects on memory and executive function, referred to as "chemo brain". It would therefore be of great value to identify biomarkers for early detection and management of this debilitating condition. TRIAL REGISTRATION Clinical Trial Registration: NCT03724994.
Collapse
Affiliation(s)
- Sofie Olsson Hau
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - Maja Svensson
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Alexandra Petersson
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jakob Eberhard
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Karin Jirström
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Barui S, Percivalle NM, Conte M, Dumontel B, Racca L, Carofiglio M, Cauda V. Development of doped ZnO-based biomimicking and tumor-targeted nanotheranostics to improve pancreatic cancer treatment. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00140-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
AbstractDespite different nanomaterials were developed so far against cancer, their potential drawbacks are still scarcely considered. The off-target delivery of a therapeutic compound, as well as the non-specific uptake of these nanomaterials by healthy tissues or organs, and their potential immunogenicity are some of the major issues that still have to be faced prior to a successful clinical translation. This work aims to develop an innovative theranostic, biocompatible, and drug-loaded nanoconstruct based on Gadolinium-doped Zinc Oxide (ZnO-Gd) nanocrystals (NCs), focusing on one of the most lethal diseases, i.e., pancreatic cancer. The use of zinc oxide is motivated by the huge potential of this nanomaterial already demonstrated for in vitro and in vivo applications, while the Gadolinium doping confers magnetic properties useful for diagnostics. Furthermore, an innovative biomimetic shell is here used to coat the NCs: it is composed of a lipid bilayer made from extracellular vesicles (EVs) combined with other synthetic lipids and a peptide targeting the pancreatic tumor microenvironment. To complete the nanoconstruct therapeutic function, Gemcitabine, a first-line drug for pancreatic cancer treatment, was adsorbed on the ZnO-Gd NCs prior to the coating with the above-mentioned lipidic shell. The aim of this work is thus to strongly enhance the therapeutic capability of the final nanoconstruct, providing it with high biocompatibility, colloidal stability in biological media, efficient cargo loading and release properties, as well as active targeting for site-selective drug delivery. Furthermore, the magnetic properties of the ZnO-Gd NCs core can in future allow efficient in situ bioimaging capabilities based on Magnetic Resonance Imaging technique. The obtained nanoconstructs were tested on two different pancreatic cancer cell lines, i.e., BxPC-3 and the metastatic AsPC-1, proving high cell internalization levels, mediated by the targeting peptide exposed on the nanoconstruct. Cellular cytotoxicity assay performed on both cell lines dictated ~ 20% increased cell killing efficacy of Gemcitabine when delivered through the nanoconstruct rather than as a free drug. Taken together, our designed theranostic nanoconstruct can have a significant impact on the standard treatment of pancreatic cancer.
Collapse
|
7
|
Kim H, Lee SH, Kim DH, Lee JY, Hong SH, Ha US, Kim IH. Gemcitabine maintenance versus observation after first-line chemotherapy in patients with metastatic urothelial carcinoma: a retrospective study. Transl Androl Urol 2020; 9:2113-2121. [PMID: 33209674 PMCID: PMC7658126 DOI: 10.21037/tau-20-772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Gemcitabine with platinum is one of the most important first-line treatments for metastatic urothelial cancer (mUC). However, continuation of platinum agents results in cumulative toxicities, such as nephrotoxicity, ototoxicity, and neurotoxicity, which lead to discontinuation of chemotherapy after 4–6 cycles despite a favorable response in the patients. The strategy of maintenance treatment can give clinical benefit to patients, but there is no consensus about maintenance treatment. The aim of this study was to investigate the clinical impact of the gemcitabine maintenance (GEM-m) in mUC patients who achieve disease control from first-line gemcitabine with platinum agents. Methods A total of 117 patients who showed response to 4–6 cycles of gemcitabine plus cisplatin or carboplatin as the first-line palliative chemotherapy were reviewed between 2014 to 2018. Patients who were treated with GEM-m received a 1,000 mg/m2 dose of gemcitabine on day 1 and 8 for 3 weeks until disease progression or development of unacceptable toxicity. The patients who are not treated with GEM-m were followed up with regular radiologic evaluation. Statistical analyses were performed using the log-rank test and Cox proportional hazards method. Results Fifty-eight patients (49.6%) received GEM-m. The median cycle of GEM-m was 4 (range, 1–12). Six patients (10.3%) in the GEM-m group showed an objective response. A median overall survival (OS) of 11.8 months and 9.6 months was observed for the GEM-m and non-GEM-m groups, respectively [HR 0.621; 95% CI, 0.39–0.97; P=0.026]. Additionally, median progression-free survival (PFS) was 4.6 months and 3.3 months in the GEM-m and non-GEM-m groups, respectively [HR 0.612; 95% CI, 0.41–0.91; P=0.009]. Grade 3 or higher neutropenia occurred in 17.2% of patients in the GEM-m and 1.7% in the non-GEM-m group. Conclusions Our results suggest that GEM-m can be considered in patients who respond to gemcitabine with platinum. Large-scale prospective study should be warranted.
Collapse
Affiliation(s)
- Hyunho Kim
- Division of Medical Oncology, Department of Internal Medicine, The Catholic University of Korea, St. Vincent's Hospital, Suwon, Republic of Korea
| | - Seung-Hwan Lee
- Division of Medical Oncology, Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Republic of Korea
| | - Dong Hwan Kim
- Department of Radiology, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Republic of Korea
| | - Ji Youl Lee
- Department of Urology Cancer Center, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Republic of Korea.,Department of Genitourinary Cancer Centre, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hoo Hong
- Department of Urology Cancer Center, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Republic of Korea.,Department of Genitourinary Cancer Centre, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - U-Syn Ha
- Department of Urology Cancer Center, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Republic of Korea.,Department of Genitourinary Cancer Centre, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Republic of Korea.,Department of Genitourinary Cancer Centre, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
8
|
Lum LG, Thakur A, Choi M, Deol A, Kondadasula V, Schalk D, Fields K, Dufrense M, Philip P, Dyson G, Aon HD, Shields AF. Clinical and immune responses to anti-CD3 x anti-EGFR bispecific antibody armed activated T cells (EGFR BATs) in pancreatic cancer patients. Oncoimmunology 2020; 9:1773201. [PMID: 32939319 PMCID: PMC7480816 DOI: 10.1080/2162402x.2020.1773201] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 01/05/2023] Open
Abstract
Purpose This was a phase I/II adoptive T cell trial in 7 locally advanced and metastatic pancreatic cancer patients using 3-8 infusions of anti-CD3 x anti-EGFR bispecific antibody armed activated T cells (BATs) to determine safety, the maximum tolerated dose (MTD), immune responses, time to progression (TTP), and overall survival (OS). Study Design: T cells obtained by apheresis were expanded and armed with EGFRBi, cryopreserved for infusions. In a phase I dose escalation, five patients received three weekly infusions of 10-40 × 109 BATs/infusion followed by a booster infusion 3 months later, and 2 patients received 8 infusions twice weekly for 4 weeks in a phase II. The trials were registered at http://www.clinicaltrials.gov, NCT01420874 and NCT02620865. Results: There were no dose-limiting toxicities (DLTs), and the targeted dose of 80 × 109 BATs was met. The median TTP is 7 months, and the median OS is 31 months. Two patients had stable disease for 6.5 and 25+ months, and two patients developed complete responses (CRs) after restarting chemotherapy. Infusions of BATs induced anti-pancreatic cancer cytotoxicity, innate immune responses, cytokine responses (IL-12, IP-10), and shifts in CD4 and CD8 Vβ repertoire with enhanced cytoplasmic IFN-γ staining in the Vβ repertoire of the CD8 subset that suggest specific clonal TCR responses. Conclusions: Infusions of BATs are safe, induce endogenous adaptive anti-tumor responses, and may have a potential to improve overall survival.
Collapse
Affiliation(s)
- Lawrence G. Lum
- Stem Cell Transplantation Program, Division of Oncology/Hematology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Archana Thakur
- Stem Cell Transplantation Program, Division of Oncology/Hematology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Minsig Choi
- Department of Oncology, Stony Brook University, Stony Brook, NY, USA
| | - Abhinav Deol
- Department of Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Vidya Kondadasula
- Department of Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Dana Schalk
- Stem Cell Transplantation Program, Division of Oncology/Hematology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Kristie Fields
- Department of Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Melissa Dufrense
- Department of Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Philip Philip
- Department of Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Gregory Dyson
- Department of Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Hussein D. Aon
- Department of Radiology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Anthony F. Shields
- Department of Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| |
Collapse
|
9
|
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]
|
10
|
Tao SF, Gu WH, Gu JC, Zhu ML, Wang Q, Zheng LZ. A Retrospective Case Series Of High-Intensity Focused Ultrasound (HIFU) In Combination With Gemcitabine And Oxaliplatin (Gemox) On Treating Elderly Middle And Advanced Pancreatic Cancer. Onco Targets Ther 2019; 12:9735-9745. [PMID: 31814733 PMCID: PMC6863124 DOI: 10.2147/ott.s220299] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/18/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose This retrospective study was conducted to evaluate the safety and efficacy of high-intensity focused ultrasound (HIFU) ablation combined with Gemcitabine and Oxaliplatin (Gemox) for the treatment of middle and advanced pancreatic cancer in elderly patients. Methods Forty-seven patients with pancreatic cancer treated with HIFU and Gemox were evaluated for inclusion, and 38 cases were finally included. The primary endpoint was safety. Secondary endpoints included the response rate, the clinical benefit response (CBR), overall survival (OS), progression-free survival (PFS). Results After combination therapy of HIFU and Gemox, severe complications were rarely reported, and no treatment-related death occurred. The rate of three or four-degree myelosuppression was low, and no obvious impairment of hepatorenal function was observed. Pancreatitis and gastrointestinal injury did not occurred. The disease control rate (DCR) was estimated to be 76.3%, including complete remission (CR), partial remission (PR), stable disease (SD) in 1, 6, 22 cases, respectively. And the objective response rate (ORR) was 18.4%. The clinical benefit rate (CBR) was 68.4%, with the pain significantly relieved (P<0.01). The serum level of CA19-9 showed significant changes after HIFU treatment. The median overall survival (OS) was 12.5 months, with a 6-month and 12-month OS rate of 82.13% and 59.34%, respectively. Stratified analyses did not reveal any significant difference between patients in different stages. Conclusion Elderly patients (≥ 60 years old) with pancreatic cancer would experience tolerable toxicity and obtain good clinical benefits from the combination therapy of HIFU ablation and Gemox.
Collapse
Affiliation(s)
- Shuang-Fen Tao
- Oncology Department, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Wen-Hua Gu
- Oncology Department, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Jian-Chun Gu
- Oncology Department, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Mei-Ling Zhu
- Oncology Department, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Qing Wang
- Oncology Department, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Lei-Zhen Zheng
- Oncology Department, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, People's Republic of China
| |
Collapse
|
11
|
Gargiulo P, Dietrich D, Herrmann R, Bodoky G, Ruhstaller T, Scheithauer W, Glimelius B, Berardi S, Pignata S, Brauchli P. Predicting mortality and adverse events in patients with advanced pancreatic cancer treated with palliative gemcitabine-based chemotherapy in a multicentre phase III randomized clinical trial: the APC-SAKK risk scores. Ther Adv Med Oncol 2019; 11:1758835918818351. [PMID: 30636977 PMCID: PMC6317152 DOI: 10.1177/1758835918818351] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022] Open
Abstract
Background The prognosis of advanced pancreatic cancer (APC) is poor and differs considerably among patients. Therefore, it is clinically relevant to identify patients with APC who are more likely to benefit from palliative chemotherapy with reduced risk of toxicity. To date, there is no prognostic score universally recommended to help clinicians in planning the therapeutic management. Methods Using individual patient data from 319 cases of APC treated with gemcitabine-based chemotherapy and enrolled in the SAKK 44/00-CECOG/PAN.1.3.001 randomized trial, several baseline variables, including inflammatory markers, were analysed post hoc as predictors of mortality and/or grade 3 or 4 chemotherapy-related toxicity and separate risk scores were developed. Results Median survival of the study patients was 7.9 months (interquartile range 3.7-13.3 months). Independent predictors of mortality included increased Aspartate transaminase (ASAT), low performance status, increased derived neutrophil to lymphocyte ratio, increased Carbohydrate Antigen 19-9 (CA 19-9), low haemoglobin, presence of pain, presence of metastasis and increased alkaline phosphatase (ALP). During the study, 117 patients experienced at least one grade 3 or 4 adverse event. Independent predictors of toxicity included white blood cells, ALP, renal function and bilirubin levels at baseline. Both models displayed moderate levels of discrimination (C-statistic 0.68 and 0.64 for mortality and toxicity, respectively) and adequate calibration. Conclusions We developed simple-to-use prognostic scores for mortality and severe toxicity for patients with APC. These scores can be useful in daily practice to identify patients with increased risk of death or toxicity and to plan the most appropriate therapeutic strategy to improve survival and quality of life. Further prospective studies to validate such scores are needed.
Collapse
Affiliation(s)
- Piera Gargiulo
- Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Effingerstrasse 33, CH-3008 Bern, Switzerland
| | - Daniel Dietrich
- Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Bern, Switzerland
| | | | | | | | | | - Bengt Glimelius
- Department of Immunology, Genetics and Pathology, University of Uppsala, Uppsala, Sweden
| | - Simona Berardi
- Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Bern, Switzerland
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori "Fondazione G. Pascale", Naples, Italy
| | - Peter Brauchli
- Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Bern, Switzerland
| |
Collapse
|
12
|
Alt-Epping B, Haas AL, Jansky M, Nauck F. Symptomlinderung durch Tumortherapie? Schmerz 2018; 32:90-98. [DOI: 10.1007/s00482-018-0270-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Chin V, Nagrial A, Sjoquist K, O'Connor CA, Chantrill L, Biankin AV, Scholten RJPM, Yip D, Cochrane Upper GI and Pancreatic Diseases Group. Chemotherapy and radiotherapy for advanced pancreatic cancer. Cochrane Database Syst Rev 2018; 3:CD011044. [PMID: 29557103 PMCID: PMC6494171 DOI: 10.1002/14651858.cd011044.pub2] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic cancer (PC) is a highly lethal disease with few effective treatment options. Over the past few decades, many anti-cancer therapies have been tested in the locally advanced and metastatic setting, with mixed results. This review attempts to synthesise all the randomised data available to help better inform patient and clinician decision-making when dealing with this difficult disease. OBJECTIVES To assess the effect of chemotherapy, radiotherapy or both for first-line treatment of advanced pancreatic cancer. Our primary outcome was overall survival, while secondary outcomes include progression-free survival, grade 3/4 adverse events, therapy response and quality of life. SEARCH METHODS We searched for published and unpublished studies in CENTRAL (searched 14 June 2017), Embase (1980 to 14 June 2017), MEDLINE (1946 to 14 June 2017) and CANCERLIT (1999 to 2002) databases. We also handsearched all relevant conference abstracts published up until 14 June 2017. SELECTION CRITERIA All randomised studies assessing overall survival outcomes in patients with advanced pancreatic ductal adenocarcinoma. Chemotherapy and radiotherapy, alone or in combination, were the eligible treatments. DATA COLLECTION AND ANALYSIS Two review authors independently analysed studies, and a third settled any disputes. We extracted data on overall survival (OS), progression-free survival (PFS), response rates, adverse events (AEs) and quality of life (QoL), and we assessed risk of bias for each study. MAIN RESULTS We included 42 studies addressing chemotherapy in 9463 patients with advanced pancreatic cancer. We did not identify any eligible studies on radiotherapy.We did not find any benefit for chemotherapy over best supportive care. However, two identified studies did not have sufficient data to be included in the analysis, and many of the chemotherapy regimens studied were outdated.Compared to gemcitabine alone, participants receiving 5FU had worse OS (HR 1.69, 95% CI 1.26 to 2.27, moderate-quality evidence), PFS (HR 1.47, 95% CI 1.12 to 1.92) and QoL. On the other hand, two studies showed FOLFIRINOX was better than gemcitabine for OS (HR 0.51 95% CI 0.43 to 0.60, moderate-quality evidence), PFS (HR 0.46, 95% CI 0.38 to 0.57) and response rates (RR 3.38, 95% CI 2.01 to 5.65), but it increased the rate of side effects. The studies evaluating CO-101, ZD9331 and exatecan did not show benefit or harm when compared with gemcitabine alone.Giving gemcitabine at a fixed dose rate improved OS (HR 0.79, 95% CI 0.66 to 0.94, high-quality evidence) but increased the rate of side effects when compared with bolus dosing.When comparing gemcitabine combinations to gemcitabine alone, gemcitabine plus platinum improved PFS (HR 0.80, 95% CI 0.68 to 0.95) and response rates (RR 1.48, 95% CI 1.11 to 1.98) but not OS (HR 0.94, 95% CI 0.81 to 1.08, low-quality evidence). The rate of side effects increased. Gemcitabine plus fluoropyrimidine improved OS (HR 0.88, 95% CI 0.81 to 0.95), PFS (HR 0.79, 95% CI 0.72 to 0.87) and response rates (RR 1.78, 95% CI 1.29 to 2.47, high-quality evidence), but it also increased side effects. Gemcitabine plus topoisomerase inhibitor did not improve survival outcomes but did increase toxicity. One study demonstrated that gemcitabine plus nab-paclitaxel improved OS (HR 0.72, 95% CI 0.62 to 0.84, high-quality evidence), PFS (HR 0.69, 95% CI 0.58 to 0.82) and response rates (RR 3.29, 95% CI 2.24 to 4.84) but increased side effects. Gemcitabine-containing multi-drug combinations (GEMOXEL or cisplatin/epirubicin/5FU/gemcitabine) improved OS (HR 0.55, 95% CI 0.39 to 0.79, low-quality evidence), PFS (HR 0.43, 95% CI 0.30 to 0.62) and QOL.We did not find any survival advantages when comparing 5FU combinations to 5FU alone. AUTHORS' CONCLUSIONS Combination chemotherapy has recently overtaken the long-standing gemcitabine as the standard of care. FOLFIRINOX and gemcitabine plus nab-paclitaxel are highly efficacious, but our analysis shows that other combination regimens also offer a benefit. Selection of the most appropriate chemotherapy for individual patients still remains difficult, with clinicopathological stratification remaining elusive. Biomarker development is essential to help rationalise treatment selection for patients.
Collapse
Affiliation(s)
- Venessa Chin
- Garvan Institute of Medical ResearchThe Kinghorn Cancer Centre384 Victoria Street DarlinghurstSydneyNSWAustralia2010
- St Vincent's HospitalSydneyNSWAustralia
| | - Adnan Nagrial
- Garvan Institute of Medical ResearchThe Kinghorn Cancer Centre384 Victoria Street DarlinghurstSydneyNSWAustralia2010
- The Crown Princess Mary Cancer CentreDarcy RoadWestmeadNSWAustralia2145
| | - Katrin Sjoquist
- University of SydneyNHMRC Clinical Trials CentreK25 ‐ Medical Foundation BuildingSydneyNSWAustralia2006
- Cancer Care Centre, St George HospitalMedical OncologySt George Hospital, Gray StKogarahAustraliaNSW 2217
| | - Chelsie A O'Connor
- St Vincent's HospitalSydneyNSWAustralia
- Genesis Cancer CareSydneyNSWAustralia
- Macquarie University HospitalSydneyAustralia
| | - Lorraine Chantrill
- The Kinghorn Cancer Centre, Garvan Institute of Medical ResearchDepartment of Pancreatic Cancer382 Victoria Street DarlinghurstSydneyNSWAustralia2010
| | - Andrew V Biankin
- University of GlasgowInstitute of Cancer SciencesWolfson Wohl Cancer Research CentreGarscube Estate, Switchback RoadGlasgowUKG61 1QH
- University of New South WalesSouth Western Sydney Clinical School, Faculty of MedicineLiverpoolNSWAustralia2170
- West of Scotland Pancreatic Unit and Glasgow Royal InfirmaryGlasgowUK
| | - Rob JPM Scholten
- Julius Center for Health Sciences and Primary Care / University Medical Center UtrechtCochrane NetherlandsRoom Str. 6.126P.O. Box 85500UtrechtNetherlands3508 GA
| | - Desmond Yip
- The Canberra HospitalDepartment of Medical OncologyYamba DriveGarranACTAustralia2605
- Australian National UniversityANU Medical SchoolActonACTAustralia0200
| | | |
Collapse
|
14
|
Zhang XW, Ma YX, Sun Y, Cao YB, Li Q, Xu CA. Gemcitabine in Combination with a Second Cytotoxic Agent in the First-Line Treatment of Locally Advanced or Metastatic Pancreatic Cancer: a Systematic Review and Meta-Analysis. Target Oncol 2018; 12:309-321. [PMID: 28353074 DOI: 10.1007/s11523-017-0486-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND It remains controversial whether the addition of a second cytotoxic agent can further improve the therapeutic effect of gemcitabine monotherapy in advanced or metastatic pancreatic cancer (LA/MPC). OBJECTIVE The objective of the present systematic review and meta-analysis was to investigate the efficacy and safety of gemcitabine-based doublet chemotherapy regimens compared to single-agent gemcitabine in the first-line treatment of unresectable LA/MPC. METHODS We searched for randomized controlled trials (RCTs) of gemcitabine monotherapy versus gemcitabine in combination with a second cytotoxic agent in patients with LA/MPC. The last search date was December 31, 2016. RESULTS Twenty-seven RCTs were identified and included in the present systematic review and meta-analysis, involving a total of 7343 patients. The meta-analysis showed that gemcitabine-based combination therapy significantly improved overall survival (OS) (HR: 0.89; 95% confidence interval (CI): 0.85-0.94; P < 0.0001), progression-free survival (PFS) (HR: 0.80; 95% CI: 0.73-0.88; P < 0.0001), and overall response rate (ORR) (RR: 1.83; 95% CI: 1.62-2.07; P < 0.0001) in comparison to single-agent gemcitabine. Subgroup analysis suggested that the antitumor activity differed between gemcitabine-based combination regimens: doublet regimens of gemcitabine plus a taxoid, and gemcitabine plus a fluoropyrimidine, in particular an oral fluoropyrimidine, resulted in a significant OS benefit for the patients. However, the combination of gemcitabine with other cytotoxic agents, such as platinum compounds or topoisomerase inhibitors failed to reduce the mortality risk. Combination therapy caused more grade 3/4 toxicities, including neutropenia, thrombocytopenia, vomiting, diarrhea, and fatigue. CONCLUSIONS Gemcitabine-based doublet regimens demonstrated superiority over gemcitabine monotherapy in overall efficacy, but were associated with increased toxicity. Different gemcitabine-based combinations showed different antitumor activity, and doublet regimens of gemcitabine in combination with a taxoid or a fluoropyrimidine, in particular an oral fluoropyrimidine provided significant survival benefits in the first-line treatment of unresectable LA/MPC.
Collapse
Affiliation(s)
- Xiu-Wei Zhang
- Department of Pathology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Yu-Xiang Ma
- Department of Oncologic Medicine, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Yang Sun
- Department of Oncologic Medicine, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Yu-Bo Cao
- Department of Oncologic Medicine, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Qin Li
- Center for Translational Medicine, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Chong-An Xu
- Department of Oncologic Medicine, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| |
Collapse
|
15
|
Determinants and prognostic value of quality of life in patients with pancreatic ductal adenocarcinoma. Eur J Cancer 2018; 92:20-32. [PMID: 29413686 DOI: 10.1016/j.ejca.2017.12.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 12/13/2017] [Accepted: 12/20/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Quality of life (QOL) is impaired in pancreatic cancer patients. Our aim was to investigate the determinants and prognostic value of QOL after diagnosis in a hospital-based cohort of racially/ethnically diverse patients with pancreatic ductal adenocarcinoma (PDAC). PATIENTS AND METHODS QOL was prospectively assessed using the Short Form-12 in 2478 PDAC patients. The Physical Component Summary (PCS) and Mental Component Summary (MCS) were categorised into tertiles based on their distribution. Ordered logistic regression was adopted to compare the risk of having lower PCS and MCS by patient sociodemographic and clinical characteristics. The association of PCS and MCS with mortality was assessed by Cox regression. RESULTS Compared with non-Hispanic whites, Hispanics were at significantly higher risk of having lower PCS (odds ratio [95% CI], 1.69 [1.26-2.26]; P < 0.001) and lower MCS (1.66 [1.24-2.23]; P < 0.001). Patients diagnosed with stage III (1.80 [1.10-2.94]; P = 0.02) and stage IV (2.32 [1.50-3.59]; P < 0.001) PDAC were more likely to have lower PCS than stage I patients. Other determinants of QOL included sex, age, drinking, smoking, education level, comorbidities and time since diagnosis. The low tertile of PCS (hazard ratio [95% CI], 1.94 [1.72-2.18]; P < 0.001) and MCS (1.42 [1.26-1.59]; P < 0.001) were each related to poor prognosis. Similar results were found for non-Hispanic whites as compared with African-Americans/Hispanics/others. CONCLUSION QOL after diagnosis is a significant prognostic indicator for patients with PDAC. Multiple factors determine QOL, suggesting possible means of intervention to improve QOL and outcomes of PDAC patients.
Collapse
|
16
|
Mané JM, Sancho A, Muñoz A, Rubio I, Fernández R, Carrera S, Fuente N, Ballesteros D, Casas R, Marrodán I, Mielgo X, López-Vivanco G. Fixed-Dose-Rate Gemcitabine Infusion in Patients with Advanced Pancreatic or Biliary Tree Adenocarcinoma. TUMORI JOURNAL 2018; 96:405-10. [DOI: 10.1177/030089161009600305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Aims and background Gemcitabine is an effective agent in pancreatic adenocarcinoma. Fixed-dose-rate gemcitabine has an interesting biological and clinical rationale, with successful results in previous studies. We conducted a trial to confirm efficacy and toxicity of fixed-dose-rate gemcitabine in patients with pancreatic or biliary tree adenocarcinoma. Methods Eligible patients with locally advanced or metastatic pancreatic or biliary tree adenocarcinoma received fixed-dose-rate gemcitabine at a dose of 1500 mg/m2 at a rate of 10 mg/m2/min weekly for 3 weeks every 28 days. Efficacy measures were overall survival, response rate and progression-free survival. Results Sixty-two patients were enrolled, and 59 were assessable for response. Seven patients (11.3%) had a partial response, 26 stable disease (41.9%) and 26 progressive disease (41.9%). Median time to progression was 21 weeks and median overall survival, 37.71 weeks. Main toxicities were grade 3–4 neutropenia (45.2%) and grade 2–3 asthenia (54.8%). No toxic deaths were documented. Conclusions Fixed-dose-rate gemcitabine has a relevant antitumor activity but with significant toxicity. It represents an interesting schedule and could be combined with other biological or chemotherapeutic agents.
Collapse
Affiliation(s)
- Joan M Mané
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| | - Aintzane Sancho
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| | - Alberto Muñoz
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| | - Itziar Rubio
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| | - Ricardo Fernández
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| | - Sergio Carrera
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| | - Natalia Fuente
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| | - David Ballesteros
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| | - Raquel Casas
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| | - Inés Marrodán
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| | - Xabier Mielgo
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| | - Guillermo López-Vivanco
- Medical Oncology Department, Hospital de Cruces, Pza Cruces s/n, Barakaldo, 48903 Bizkaia, Spain
| |
Collapse
|
17
|
Matera R, Saif MW. New therapeutic directions for advanced pancreatic cancer: cell cycle inhibitors, stromal modifiers and conjugated therapies. Expert Opin Emerg Drugs 2017; 22:223-233. [PMID: 28783977 DOI: 10.1080/14728214.2017.1362388] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Pancreatic adenocarcinoma is a devastating malignancy with an extremely poor prognosis. These tumors progress rapidly and somewhat silently with few specific symptoms and are relatively resistant to chemotherapeutic agents. Many agents, including cell cycle inhibitors, are under development for the treatment of this cancer for which there are disappointingly few treatment options. Areas covered: Here we outline the existing approved treatments for advanced pancreatic disease and discuss a range of novel therapies currently under development including cell cycle inhibitors, stromal modifiers and conjugated therapies. We also describe the current state of the pancreatic cancer therapeutics market both past and future. Expert opinion: Despite the recent explosion of novel therapies with an array of unique targets, the core treatment of pancreatic cancer still with traditional cytotoxic agents with a few exceptions. However, as these novel treatments move through the pipeline, we are hopeful that there will soon be a number of effective options for patients with advanced pancreatic cancer.
Collapse
Affiliation(s)
- Robert Matera
- a Department of Hematology and Oncology , Tufts University School of Medicine , Boston , MA , USA
| | - Muhammad Wasif Saif
- a Department of Hematology and Oncology , Tufts University School of Medicine , Boston , MA , USA
| |
Collapse
|
18
|
Borsoi C, Leonard F, Lee Y, Zaid M, Elganainy D, Alexander JF, Kai M, Liu YT, Kang Y, Liu X, Koay EJ, Ferrari M, Godin B, Yokoi K. Gemcitabine enhances the transport of nanovector-albumin-bound paclitaxel in gemcitabine-resistant pancreatic ductal adenocarcinoma. Cancer Lett 2017; 403:296-304. [PMID: 28687352 DOI: 10.1016/j.canlet.2017.06.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/03/2017] [Accepted: 06/16/2017] [Indexed: 02/06/2023]
Abstract
The mechanism for improved therapeutic efficacy of the combination therapy with nanoparticle albumin-bound paclitaxel (nAb-PTX) and gemcitabine (gem) for pancreatic ductal adenocarcinoma (PDAC) has been ascribed to enhanced gem transport by nAb-PTX. Here, we used an orthotopic mouse model of gem-resistant human PDAC in which increasing gem transport would not improve the efficacy, thus revealing the importance of nAb-PTX transport. We aimed to evaluate therapeutic outcomes and transport of nAb-PTX to PDAC as a result of (1) encapsulating nAb-PTX in multistage nanovectors (MSV); (2) effect of gem on caveolin-1 expression. Treatment with MSV/nAb-PTX + gem was highly efficient in prolonging animal survival in comparison to other therapeutic regimens. MSV/nAb-PTX + gem also caused a substantial increase in tumor PTX accumulation, significantly reduced tumor growth and tumor cell proliferation, and increased apoptosis. Moreover, gem enhanced caveolin-1 expression in vitro and in vivo, thereby improving transport of nAb-PTX to PDAC. This data was confirmed by analysis of PDACs from patients who received gem-based neo-adjuvant chemotherapy. In conclusion, we found that nAb-PTX treatment of gem-resistant PDAC can be enhanced by (1) gem through up-regulation of caveolin-1 and (2) MSV through increasing accumulation of nAb-PTX in the tumor.
Collapse
Affiliation(s)
- Carlotta Borsoi
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Fransisca Leonard
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Yeonju Lee
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Mohamed Zaid
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Dalia Elganainy
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | | | - Megumi Kai
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Yan Ting Liu
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Yaan Kang
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA
| | - Eugene J Koay
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA.
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA.
| | - Kenji Yokoi
- Department of Nanomedicine, Houston Methodist Research Institute, 6700 Bertner Ave., Houston, TX 77030, USA.
| |
Collapse
|
19
|
Khalafalla FG, Khan MW. Inflammation and Epithelial-Mesenchymal Transition in Pancreatic Ductal Adenocarcinoma: Fighting Against Multiple Opponents. CANCER GROWTH AND METASTASIS 2017; 10:1179064417709287. [PMID: 28579826 PMCID: PMC5436837 DOI: 10.1177/1179064417709287] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and one of the most lethal human cancers. Inflammation is a critical component in PDAC initiation and progression. Inflammation also contributes to the aggressiveness of PDAC indirectly via induction of epithelial-mesenchymal transition (EMT), altogether leading to enhanced resistance to chemotherapy and poor survival rates. This review gives an overview of the key pro-inflammatory signaling pathways involved in PDAC pathogenesis and discusses the role of inflammation in induction of EMT and development of chemoresistance in patients with PDAC.
Collapse
|
20
|
Hagiwara Y, Ohashi Y, Okusaka T, Ueno H, Ioka T, Boku N, Egawa S, Hatori T, Furuse J, Mizumoto K, Ohkawa S, Yamaguchi T, Yamao K, Funakoshi A, Cheng AL, Kihara K, Sato A, Tanaka M. Health-related quality of life in a randomised phase III study of gemcitabine plus S-1, S-1 alone and gemcitabine alone for locally advanced or metastatic pancreatic cancer: GEST study. ESMO Open 2017; 2:e000151. [PMID: 28761731 PMCID: PMC5519786 DOI: 10.1136/esmoopen-2016-000151] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 12/14/2022] Open
Abstract
Objective: This study was performed to compare health-related quality of life (HRQOL) of gemcitabine plus S-1 (GS), S-1 alone and gemcitabine alone as first-line chemotherapy for locally advanced or metastatic pancreatic cancer in the GEST (Gemcitabine and TS-1 Trial) study and to assess the impacts of adverse events and tumour response on HRQOL. Methods: Patients were randomly assigned to receive gemcitabine alone (1000 mg/m2 weekly for 3 of 4 weeks), S-1 alone (80, 100 or 120 mg/day twice daily for 4 of 6 weeks) or GS (gemcitabine at 1000 mg/m2 weekly plus S-1 at 60, 80 or 100 mg/day twice daily for 2 of 3 weeks). HRQOL was assessed using the EuroQoL-5D (EQ-5D) questionnaire at baseline and weeks 6, 12, 24, 48 and 72. EQ-5D scores, quality-adjusted life months (QALMs), quality-adjusted progression-free months (QAPFMs) and time until definitive HRQOL deterioration (TUDD) were compared among the three groups. The impacts of adverse events and tumour response on EQ-5D scores were analysed. Results: Including EQ-5D scores after death as 0, the mean profile was significantly better in the GS than gemcitabine group (difference, 0.069; p=0.003), but not the S-1 group (difference, −0.011; p=0.613). The mean profiles until death were similar in the three groups. QALMs, QAPFMs and TUDD were significantly longer in the GS than gemcitabine group (p<0.001, p<0.001 and p=0.004, respectively), but not the S-1 group (p=0.563, p=0.741 and p=0.701, respectively). Fatigue, anorexia and tumour response were significantly associated with changes in EQ-5D scores. Conclusions: GS achieved better HRQOL than gemcitabine alone, resulting a good balance between overall survival and HRQOL benefits. S-1 alone provides HRQOL similar to that provided by gemcitabine alone. Preventing fatigue and anorexia and maintaining better response would improve HRQOL.
Collapse
Affiliation(s)
| | - Yasuo Ohashi
- Department of Integrated Science and Engineering for Sustainable Society, Chuo University, Tokyo, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hideki Ueno
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsuya Ioka
- Department of Hepatobiliary and Pancreatic Oncology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Narikazu Boku
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | | | - Takashi Hatori
- Digestive Diseases Center, International University of Health and Welfare Mita Hospital, Tokyo, Japan
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University School of Medicine, Tokyo, Japan
| | | | - Shinichi Ohkawa
- Division of Hepatobiliary and Pancreatic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Taketo Yamaguchi
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Kenji Yamao
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | | | - Ann-Lii Cheng
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Kiyohiro Kihara
- Department of Biostatistics, The University of Tokyo, Tokyo, Japan
| | - Atsushi Sato
- Department of Medical Oncology, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Masao Tanaka
- Department of Surgery, Shimonoseki City Hospital, Shimonoseki, Japan
| |
Collapse
|
21
|
Balaban EP, Mangu PB, Khorana AA, Shah MA, Mukherjee S, Crane CH, Javle MM, Eads JR, Allen P, Ko AH, Engebretson A, Herman JM, Strickler JH, Benson AB, Urba S, Yee NS. Locally Advanced, Unresectable Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2016; 34:2654-2668. [PMID: 27247216 DOI: 10.1200/jco.2016.67.5561] [Citation(s) in RCA: 271] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To provide evidence-based recommendations to oncologists and others for treatment of patients with locally advanced, unresectable pancreatic cancer. METHODS American Society of Clinical Oncology convened an Expert Panel of medical oncology, radiation oncology, surgical oncology, gastroenterology, palliative care, and advocacy experts and conducted a systematic review of the literature from January 2002 to June 2015. Outcomes included overall survival, disease-free survival, progression-free survival, and adverse events. RESULTS Twenty-six randomized controlled trials met the systematic review criteria. RECOMMENDATIONS A multiphase computed tomography scan of the chest, abdomen, and pelvis should be performed. Baseline performance status and comorbidity profile should be evaluated. The goals of care, patient preferences, psychological status, support systems, and symptoms should guide decisions for treatments. A palliative care referral should occur at first visit. Initial systemic chemotherapy (6 months) with a combination regimen is recommended for most patients (for some patients radiation therapy may be offered up front) with Eastern Cooperative Oncology Group performance status 0 or 1 and a favorable comorbidity profile. There is no clear evidence to support one regimen over another. The gemcitabine-based combinations and treatments recommended in the metastatic setting (eg, fluorouracil, leucovorin, irinotecan, and oxaliplatin and gemcitabine plus nanoparticle albumin-bound paclitaxel) have not been evaluated in randomized controlled trials involving locally advanced, unresectable pancreatic cancer. If there is local disease progression after induction chemotherapy, without metastasis, then radiation therapy or stereotactic body radiotherapy may be offered also with an Eastern Cooperative Oncology Group performance status ≤ 2 and an adequate comorbidity profile. If there is stable disease after 6 months of induction chemotherapy but unacceptable toxicities, radiation therapy may be offered as an alternative. Patients with disease progression should be offered treatment per the ASCO Metastatic Pancreatic Cancer Treatment Guideline. Follow-up visits every 3 to 4 months are recommended. Additional information is available at www.asco.org/guidelines/LAPC and www.asco.org/guidelines/MetPC and www.asco.org/guidelineswiki.
Collapse
Affiliation(s)
- Edward P Balaban
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Pamela B Mangu
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Alok A Khorana
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Manish A Shah
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Somnath Mukherjee
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Christopher H Crane
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Milind M Javle
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Jennifer R Eads
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Peter Allen
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Andrew H Ko
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Anitra Engebretson
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Joseph M Herman
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - John H Strickler
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Al B Benson
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Susan Urba
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Nelson S Yee
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| |
Collapse
|
22
|
Arshad A, Isherwood J, Mann C, Cooke J, Pollard C, Runau F, Morgan B, Steward W, Metcalfe M, Dennison A. Intravenous ω-3 Fatty Acids Plus Gemcitabine. JPEN J Parenter Enteral Nutr 2016. [PMID: 26220200 DOI: 10.1177/0148607115595221] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Marine-derived ω-3 fatty acids (ω-3FAs) have proven antitumor activity in vivo and in vitro and improve quality of life (QOL) in clinical cancer studies. These changes may be mediated by reduction in circulating proangiogenic and proinflammatory factors. In this first study of intravenous ω-3FAs as a therapy in cancer patients, we aimed to assess if it could augment the antitumor activity of gemcitabine in patients with advanced pancreatic cancer and improve QOL. MATERIALS AND METHODS Patients were administered gemcitabine 1000 mg/m3 weekly followed by up to 100 g (200 mg/mL) of ω-3 rich lipid emulsion for 3 weeks followed by a rest week. This was continued for up to 6 cycles, progression, unacceptable toxicity, patient request, or death. The primary outcome measure was objective response rate, with secondary outcome measures of overall and progression free survival, QOL scores, and adverse events. RESULTS Fifty patients were recruited. Response rate was 14.3% and disease control rate was 85.7%. Overall and progression free survival were 5.9 and 4.8 months, respectively. Increase in global health of > 10% over baseline was seen in 47.2% of patients. More than 50% of patients had > 10% increase in QOL scores in generic symptom scores and both disease-specific domains. Grade 3/4 adverse events were thrombocytopenia (8%), neutropenia (12%), nausea or vomiting (4%), and chills (6%). CONCLUSION Intravenous ω-3FAs in combination with gemcitabine shows evidence of improved activity and benefit to QOL in patients with advanced pancreas cancer and is worthy of investigation in a randomized phase III trial.
Collapse
Affiliation(s)
- Ali Arshad
- 1 Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester, Leicester, UK
| | - John Isherwood
- 1 Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester, Leicester, UK
| | - Christopher Mann
- 1 Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester, Leicester, UK
| | - Jill Cooke
- 1 Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester, Leicester, UK
| | - Cristina Pollard
- 1 Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester, Leicester, UK
| | - Franscois Runau
- 1 Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester, Leicester, UK
| | - Bruno Morgan
- 2 Department of Radiology, University Hospitals of Leicester, Leicester, UK
| | - William Steward
- 3 Department of Medical Oncology, University Hospitals of Leicester, Leicester, UK
| | - Matthew Metcalfe
- 1 Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester, Leicester, UK
| | - Ashley Dennison
- 1 Department of Hepatobiliary and Pancreatic Surgery, University Hospitals of Leicester, Leicester, UK
| |
Collapse
|
23
|
Sohal DPS, Mangu PB, Khorana AA, Shah MA, Philip PA, O'Reilly EM, Uronis HE, Ramanathan RK, Crane CH, Engebretson A, Ruggiero JT, Copur MS, Lau M, Urba S, Laheru D. Metastatic Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2016; 34:2784-96. [PMID: 27247222 DOI: 10.1200/jco.2016.67.1412] [Citation(s) in RCA: 229] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To provide evidence-based recommendations to oncologists and others for the treatment of patients with metastatic pancreatic cancer. METHODS American Society of Clinical Oncology convened an Expert Panel of medical oncology, radiation oncology, surgical oncology, gastroenterology, palliative care, and advocacy experts to conduct a systematic review of the literature from April 2004 to June 2015. Outcomes were overall survival, disease-free survival, progression-free survival, and adverse events. RESULTS Twenty-four randomized controlled trials met the systematic review criteria. RECOMMENDATIONS A multiphase computed tomography scan of the chest, abdomen, and pelvis should be performed. Baseline performance status and comorbidity profile should be evaluated. Goals of care, patient preferences, treatment response, psychological status, support systems, and symptom burden should guide decisions for treatments. A palliative care referral should occur at first visit. FOLFIRINOX (leucovorin, fluorouracil, irinotecan, and oxaliplatin; favorable comorbidity profile) or gemcitabine plus nanoparticle albumin-bound (NAB) -paclitaxel (adequate comorbidity profile) should be offered to patients with Eastern Cooperative Oncology Group performance status (ECOG PS) 0 to 1 based on patient preference and support system available. Gemcitabine alone is recommended for patients with ECOG PS 2 or with a comorbidity profile that precludes other regimens; the addition of capecitabine or erlotinib may be offered. Patients with an ECOG PS ≥ 3 and poorly controlled comorbid conditions should be offered cancer-directed therapy only on a case-by-case basis; supportive care should be emphasized. For second-line therapy, gemcitabine plus NAB-paclitaxel should be offered to patients with first-line treatment with FOLFIRINOX, an ECOG PS 0 to 1, and a favorable comorbidity profile; fluorouracil plus oxaliplatin, irinotecan, or nanoliposomal irinotecan should be offered to patients with first-line treatment with gemcitabine plus NAB-paclitaxel, ECOG PS 0 to 1, and favorable comorbidity profile, and gemcitabine or fluorouracil should be offered to patients with either an ECOG PS 2 or a comorbidity profile that precludes other regimens. Additional information is available at www.asco.org/guidelines/MetPC and www.asco.org/guidelineswiki.
Collapse
Affiliation(s)
- Davendra P S Sohal
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Pamela B Mangu
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Alok A Khorana
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Manish A Shah
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Philip A Philip
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Eileen M O'Reilly
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Hope E Uronis
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Ramesh K Ramanathan
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Christopher H Crane
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Anitra Engebretson
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Joseph T Ruggiero
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Mehmet S Copur
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Michelle Lau
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Susan Urba
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Daniel Laheru
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| |
Collapse
|
24
|
Kristensen A, Vagnildhaug OM, Grønberg BH, Kaasa S, Laird B, Solheim TS. Does chemotherapy improve health-related quality of life in advanced pancreatic cancer? A systematic review. Crit Rev Oncol Hematol 2016; 99:286-98. [PMID: 26819138 DOI: 10.1016/j.critrevonc.2016.01.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 12/18/2015] [Accepted: 01/12/2016] [Indexed: 01/05/2023] Open
Abstract
Chemotherapy is increasingly being used in advanced pancreatic cancer, but side-effects are common. The aim of this systematic review was to assess whether chemotherapy improves health-related quality of life (HRQoL), pain or cachexia. Thirty studies were reviewed. Four of 23 studies evaluating HRQoL, 7 of 24 studies evaluating pain and 0 of 8 studies evaluating cachexia found differences between treatment arms. Change in HRQoL from baseline was evaluated in 14 studies: five studies reported an improvement in at least one treatment arm; three a worsening and the remaining stable scores. Change in pain intensity from baseline was evaluated in eight studies, and improvement was observed in seven. Of the four studies reporting improved survival, three reported improved HRQoL or pain. In conclusion, chemotherapy can stabilize HRQoL and improve pain control. Effects on cachexia are hard to elucidate. Improved survival does not come at the expense of HRQoL or pain control.
Collapse
Affiliation(s)
- A Kristensen
- European Palliative Care Research Centre (PRC), Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway; The Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| | - O M Vagnildhaug
- European Palliative Care Research Centre (PRC), Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway; The Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - B H Grønberg
- European Palliative Care Research Centre (PRC), Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway; The Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - S Kaasa
- European Palliative Care Research Centre (PRC), Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway; The Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - B Laird
- European Palliative Care Research Centre (PRC), Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway; University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - T S Solheim
- European Palliative Care Research Centre (PRC), Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway; The Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
25
|
Mao YH, Yu CZ. Current situation and predicament of multidisciplinary treatment of pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2015; 23:5750-5759. [DOI: 10.11569/wcjd.v23.i36.5750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic adenocarcinoma is one of the deadliest solid malignancies. As the anatomy position of the pancreas is deep, most patients are difficult to diagnose in the early stage. Approximately 40% of patients are diagnosed with locally advanced or metastatic disease at the time of diagnosis, which severely limits the number of patients who can undergo surgical resection. There is an urgent need to explore new treatments or comprehensive treatment for pancreatic cancer, in order to improve the long-term survival rate and quality of life. Recent therapeutic advances for advanced pancreatic cancer have improved overall survival, but the prognosis remains grim. This article reviews both the major therapeutic strategies and their predicaments for pancreatic adenocarcinoma, including currently ongoing clinical trials about use of the new agents and technologies.
Collapse
|
26
|
Tu C, Zheng F, Wang JY, Li YY, Qian KQ. An Updated Meta-analysis and System Review:is Gemcitabine+Fluoropyrimidine in Combination a Better Therapy Versus Gemcitabine Alone for Advanced and Unresectable Pancreatic Cancer? Asian Pac J Cancer Prev 2015; 16:5681-6. [DOI: 10.7314/apjcp.2015.16.14.5681] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
27
|
Young SJ. Irreversible electroporation and the pancreas: What we know and where we are going? World J Gastrointest Surg 2015; 7:138-144. [PMID: 26328033 PMCID: PMC4550840 DOI: 10.4240/wjgs.v7.i8.138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 06/02/2015] [Accepted: 07/02/2015] [Indexed: 02/06/2023] Open
Abstract
Pancreatic adenocarcinoma continues to have a poor prognosis with 1 and 5 years survival rates of 27% and 6% respectively. The gold standard of treatment is resection, however, only approximately 10% of patients present with resectable disease. Approximately 40% of patients present with disease that is too locally advanced to resect. There is great interest in improving outcomes in this patient population and ablation techniques have been investigated as a potential solution. Unfortunately early investigations into thermal ablation techniques, particularly radiofrequency ablation, resulted in unacceptably high morbidity rates. Irreversible electroporation (IRE) has been introduced and is promising as it does not rely on thermal energy and has shown an ability to leave structural cells such as blood vessels and bile ducts intact during animal studies. IRE also does not suffer from heat sink effect, a concern given the large number of blood vessels surrounding the pancreas. IRE showed significant promise during preclinical animal trials and as such has moved on to clinical testing. There are as of yet only a few studies which look at the applications of IRE within humans in the setting of pancreatic adenocarcinoma. This paper reviews the basic principles, techniques, and current clinical data available on IRE.
Collapse
|
28
|
Namba T, Kodama R, Moritomo S, Hoshino T, Mizushima T. Zidovudine, an anti-viral drug, resensitizes gemcitabine-resistant pancreatic cancer cells to gemcitabine by inhibition of the Akt-GSK3β-Snail pathway. Cell Death Dis 2015; 6:e1795. [PMID: 26111057 PMCID: PMC4669843 DOI: 10.1038/cddis.2015.172] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/11/2015] [Accepted: 05/27/2015] [Indexed: 12/11/2022]
Abstract
Pancreatic cancer is one of the most difficult malignancies to treat owing to the rapid acquisition of resistance to chemotherapy. Gemcitabine, a first-line treatment for pancreatic cancer, prolongs patient survival by several months, and combination treatment with gemcitabine and other anti-cancer drugs in the clinic do not show any significant effects on overall survival. Thus, identification of a drug that resensitizes gemcitabine-resistant pancreatic cancer to gemcitabine and a better understanding of the molecular mechanisms of gemcitabine resistance are critical to develop new therapeutic options for pancreatic cancer. Here, we report that zidovudine resensitizes gemcitabine-resistant pancreatic cancer to gemcitabine as shown by screening a compound library, including clinical medicine, using gemcitabine-resistant cells. In analyzing the molecular mechanisms of zidovudine effects, we found that the epithelial-to-mesenchymal transition (EMT)-like phenotype and downregulation of human equilibrative nucleoside transporter 1 (hENT1) are essential for the acquisition of gemcitabine resistance, and zidovudine restored these changes. The chemical biology investigations also revealed that activation of the Akt-GSK3β-Snail1 pathway in resistant cells is a key signaling event for gemcitabine resistance, and zidovudine resensitized resistant cells to gemcitabine by inhibiting this activated pathway. Moreover, our in vivo study demonstrated that co-administration of zidovudine and gemcitabine strongly suppressed the formation of tumors by gemcitabine-resistant pancreatic cancer and prevented gemcitabine-sensitive pancreatic tumors from acquiring gemcitabine-resistant properties, inducing an EMT-like phenotype and downregulating hENT1 expression. These results suggested that co-treatment with zidovudine and gemcitabine may become a novel therapeutic strategy for pancreatic cancer by inhibiting chemoresistance-specific signaling.
Collapse
Affiliation(s)
- T Namba
- Science Research Center, Kochi University, Kochi 783-8505, Japan
| | - R Kodama
- Science Research Center, Kochi University, Kochi 783-8505, Japan
| | - S Moritomo
- Graduate School of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - T Hoshino
- Department of Analytical Chemistry, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - T Mizushima
- Department of Analytical Chemistry, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| |
Collapse
|
29
|
Anota A, Mouillet G, Trouilloud I, Dupont-Gossart AC, Artru P, Lecomte T, Zaanan A, Gauthier M, Fein F, Dubreuil O, Paget-Bailly S, Taieb J, Bonnetain F. Sequential FOLFIRI.3 + Gemcitabine Improves Health-Related Quality of Life Deterioration-Free Survival of Patients with Metastatic Pancreatic Adenocarcinoma: A Randomized Phase II Trial. PLoS One 2015; 10:e0125350. [PMID: 26010884 PMCID: PMC4444351 DOI: 10.1371/journal.pone.0125350] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 03/19/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND A randomized multicenter phase II trial was conducted to assess the sequential treatment strategy using FOLFIRI.3 and gemcitabine alternately (Arm 2) compared to gemcitabine alone (Arm 1) in patients with metastatic non pre-treated pancreatic adenocarcinoma. The primary endpoint was the progression-free survival (PFS) rate at 6 months. It concludes that the sequential treatment strategy appears to be feasible and effective with a PFS rate of 43.5% in Arm 2 at 6 months (26.1% in Arm 1). This paper reports the results of the longitudinal analysis of the health-related quality of life (HRQoL) as a secondary endpoint of this study. METHODS HRQoL was evaluated using the EORTC QLQ-C30 at baseline and every two months until the end of the study or death. HRQoL deterioration-free survival (QFS) was defined as the time from randomization to a first significant deterioration as compared to the baseline score with no further significant improvement, or death. A propensity score was estimated comparing characteristics of partial and complete responders. Analyses were repeated with inverse probability weighting method using the propensity score. Multivariate Cox regression analyses were performed to identify independent factors influencing QFS. RESULTS 98 patients were included between 2007 and 2011. Adjusting on the propensity score, patients of Arm 2 presented a longer QFS of Global Health Status (Hazard Ratio: 0.52 [0.31-0.85]), emotional functioning (0.35 [0.21-0.59]) and pain (0.50 [0.31-0.81]) than those of Arm 1. CONCLUSION Patients of Arm 2 presented a better HRQoL with a longer QFS than those of Arm 1. Moreover, the propensity score method allows to take into account the missing data depending on patients' characteristics. TRIAL REGISTRATION INFORMATION Eudract N° 2006-005703-34. (Name of the Trial: FIRGEM).
Collapse
Affiliation(s)
- Amélie Anota
- National Quality of Life in Oncology Platform, Besançon, France
- Methodological and Quality of Life in Oncology Unit (EA 3181), University Hospital of Besançon, Besançon, France
| | - Guillaume Mouillet
- Methodological and Quality of Life in Oncology Unit (EA 3181), University Hospital of Besançon, Besançon, France
| | - Isabelle Trouilloud
- Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, University of Paris Descartes, Paris, France
| | | | - Pascal Artru
- Hepato-Gastro-Enterology and Digestive Oncology Department, Hospital Jean Mermoz, Lyon, France
| | - Thierry Lecomte
- Department of Gastroenterology and Digestive Oncology, University Hospital of Tours- Trousseau Hospital, Chambray-Les-Tours, France
| | - Aziz Zaanan
- Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, University of Paris Descartes, Paris, France
| | - Mélanie Gauthier
- Biostatistics and Quality of Life Unit, Centre Georges François Leclerc, Dijon, France
| | - Francine Fein
- Department of Gastroenterology, University Hospital of Besançon, Besançon, France
| | - Olivier Dubreuil
- Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, University of Paris Descartes, Paris, France
| | - Sophie Paget-Bailly
- Methodological and Quality of Life in Oncology Unit (EA 3181), University Hospital of Besançon, Besançon, France
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, University of Paris Descartes, Paris, France
| | - Franck Bonnetain
- National Quality of Life in Oncology Platform, Besançon, France
- Methodological and Quality of Life in Oncology Unit (EA 3181), University Hospital of Besançon, Besançon, France
| |
Collapse
|
30
|
Collins DC, Morris PG. Systemic therapy for advanced pancreatic cancer: individualising cytotoxic therapy. Expert Opin Pharmacother 2015; 16:851-61. [DOI: 10.1517/14656566.2015.1024654] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
31
|
Patient-reported outcomes as end points and outcome indicators in solid tumours. Nat Rev Clin Oncol 2015; 12:358-70. [DOI: 10.1038/nrclinonc.2015.29] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
32
|
Teague A, Lim KH, Wang-Gillam A. Advanced pancreatic adenocarcinoma: a review of current treatment strategies and developing therapies. Ther Adv Med Oncol 2015; 7:68-84. [PMID: 25755680 DOI: 10.1177/1758834014564775] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pancreatic adenocarcinoma is one of the deadliest solid malignancies. A large proportion of patients are diagnosed with locally advanced or metastatic disease at the time of presentation and, unfortunately, this severely limits the number of patients who can undergo surgical resection, which offers the only chance for cure. Recent therapeutic advances for patients with advanced pancreatic cancer have extended overall survival, but prognosis still remains grim. Given that traditional chemotherapy is ineffective in curing advanced pancreatic adenocarcinoma, current research is taking a multidirectional approach in the hopes of developing more effective treatments. This article reviews the major clinical trial data that is the basis for the current chemotherapy regimens used as first- and second-line treatments for advanced pancreatic adenocarcinoma. We also review the current ongoing clinical trials, which include the use of agents targeting the oncogenic network signaling of K-Ras, agents targeting the extracellular matrix, and immune therapies.
Collapse
Affiliation(s)
- Andrea Teague
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrea Wang-Gillam
- Division of Oncology, Department of Medicine, Campus Box 8056, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
33
|
Sherman WH, Chu K, Chabot J, Allendorf J, Schrope BA, Hecht E, Jin B, Leung D, Remotti H, Addeo G, Postolov I, Tsai W, Fine RL. Neoadjuvant gemcitabine, docetaxel, and capecitabine followed by gemcitabine and capecitabine/radiation therapy and surgery in locally advanced, unresectable pancreatic adenocarcinoma. Cancer 2015; 121:673-80. [PMID: 25492104 DOI: 10.1002/cncr.29112] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 09/07/2014] [Accepted: 09/08/2014] [Indexed: 01/11/2023]
Abstract
BACKGROUND This prospective study was undertaken to assess toxicity, resectability, and survival in pancreatic adenocarcinoma patients presenting with locally advanced, unresectable disease treated with neoadjuvant gemcitabine, docetaxel, and capecitabine (GTX) and gemcitabine and capecitabine (GX)/radiation therapy (RT). METHODS All patients presenting to the Pancreas Center were evaluated for eligibility. Forty-five patients (mean age, 64 years; range, 44-83 years)-34 patients deemed unresectable because of arterial involvement and 11 patients deemed unresectable because of extensive venous involvement-were treated with 6 cycles of GTX. Those with arterial involvement were treated with GX/RT after chemotherapy. RESULTS The GTX and GX/RT treatments were tolerated with the expected drug-related toxicities. There were no bowel perforations, cases of pancreatitis, or delayed strictures. Among those with arterial involvement, 29 underwent subsequent resection, with 20 (69%) achieving R0 resections. All 11 patients with venous-only involvement underwent resection, with 8 achieving R0 resections and 3 achieving complete pathologic responses. For the arterial arm, the 1-year survival rate was 71% (24 of 34 patients), and the median survival was 29 months (95% confidence interval, 21-38 months). Thirteen patients (38%) have not relapsed (range, 5-49+ months). For the venous arm, the median survival has not been reached at more than 42 months. Six patients (55%) in the venous arm did not experience recurrence (range, 6.2-42+ months). CONCLUSIONS GTX plus GX/RT is an effective neoadjuvant regimen that can be safely administered to patients up to at least the age of 83 years. It is associated with a high response rate, a high rate of R0 resections, and prolonged overall survival.
Collapse
|
34
|
Pancreatic cancer: diagnosis and treatments. Tumour Biol 2015; 36:1375-84. [PMID: 25680410 DOI: 10.1007/s13277-015-3223-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 02/03/2015] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is one of the deadliest cancers, with exceptionally high mortality. Despite the relatively low incidence rate (10th), it is the fourth leading cause of cancer-related deaths in most developed countries. To improve the early diagnosis of pancreatic cancer and strengthen the standardized comprehensive treatment are still the main focus of pancreatic cancer research. Here, we summarized the rapid developments in the diagnosis and treatments of pancreatic cancer. Regarding diagnosis, we reviewed advances in medical imaging technology, tumor markers, molecular biology (e.g., gene mutation), and proteomics. Moreover, great progress has also been made in the treatments of this disease, including surgical resection, chemotherapy, targeted radiotherapy, targeted minimally invasive treatment, and molecular targeted therapy. Therefore, we also recapitulated the development, advantages, and disadvantages of each of the treatment methods in this review.
Collapse
|
35
|
Wang C, Zhang W, Fu M, Yang A, Huang H, Xie J. Establishment of human pancreatic cancer gemcitabine‑resistant cell line with ribonucleotide reductase overexpression. Oncol Rep 2014; 33:383-90. [PMID: 25394408 DOI: 10.3892/or.2014.3599] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 08/29/2014] [Indexed: 12/12/2022] Open
Abstract
A gemcitabine (GEM)-resistant human pancreatic cancer cell line (PANC-1RG7) was established in vitro by gradually increasing GEM concentrations and cloning cell cultures to develop a cellular model of acquired drug resistance studies. We found that PANC-1RG7 cells exhibited significantly different morphological characteristics from parental cells. PANC-1RG7 cells grew slowly (p<0.05), yet the cell cycle remained unchanged (p>0.05). PANC-1RG7, with a resistance index to GEM of 39.9, showed cross-resistance characteristics to methotrexate, gefitinib, cisplatin and 5-fluorouracil. The proliferation inhibition of GEM was significantly reduced in vivo (p<0.05). The known resistance-associated genes and proteins we detected remained unchanged, with the exception of cytidine deaminase, multidrug resistance-related protein and breast cancer resistance protein genes, which decreased; by contrast, 5'-nucleotidase, ribonucleotide reductase (RRM) 1 and RRM2 proteins increased (p<0.05). Therefore, a cell line with acquired GEM resistance was established successfully. Resistance was acquired by overexpressing RRM1 and RRM2 proteins.
Collapse
Affiliation(s)
- Congfei Wang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Weiwei Zhang
- Department of Pharmacology, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Mingjuan Fu
- Department of Pharmacology, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Aiqin Yang
- Department of Pharmacology, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jieming Xie
- Department of Pharmacology, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| |
Collapse
|
36
|
Serrano PE, Herman JM, Griffith KA, Zalupski MM, Kim EJ, Bekaii-Saab TS, Ben-Josef E, Dawson LA, Ringash J, Wei AC. Quality of life in a prospective, multicenter phase 2 trial of neoadjuvant full-dose gemcitabine, oxaliplatin, and radiation in patients with resectable or borderline resectable pancreatic adenocarcinoma. Int J Radiat Oncol Biol Phys 2014; 90:270-7. [PMID: 25104069 DOI: 10.1016/j.ijrobp.2014.05.053] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/05/2014] [Accepted: 05/27/2014] [Indexed: 12/30/2022]
Abstract
PURPOSE To determine the health-related quality of life (QOL) during and after neoadjuvant chemoradiation therapy and surgery for patients with pancreatic adenocarcinoma. METHODS AND MATERIALS Participants of a prospective, phase 2 multi-institutional trial treated with neoadjuvant chemoradiation followed by surgery completed QOL questionnaires (European Organization for Research and Treatment in Cancer Quality of Life Questionnaire version 3.0 [EORTC-QLQ C30], EORTC-Pancreatic Cancer module [EORTC-PAN 26], and Functional Assessment of Cancer Therapy Hepatobiliary and Pancreatic subscale [FACT-Hep]) at baseline, after 2 cycles of neoadjuvant therapy, after surgery, at 6 months from initiation of therapy, and at 6-month intervals for 2 years. Mean scores were compared with baseline. A change >10% was considered a minimal clinically important difference. RESULTS Of 71 participants in the trial, 55 were eligible for QOL analysis. Compliance ranged from 32% to 74%. The EORTC-QLQ C30 global QOL did not significantly decline after neoadjuvant therapy, whereas the Functional Assessment of Cancer Therapy global health measure showed a statistically, but not clinically significant decline (-8, P=.02). This was in parallel with deterioration in physical functioning (-14.1, P=.001), increase in diarrhea (+16.7, P=.044), and an improvement in pancreatic pain (-13, P=.01) as per EORTC-PAN 26. Because of poor patient compliance in the nonsurgical group, long-term analysis was performed only from surgically resected participants (n=36). Among those, global QOL returned to baseline levels after 6 months, remaining near baseline through the 24-month visit. CONCLUSIONS The study regimen consisting of 2 cycles of neoadjuvant therapy was completed without a clinically significant QOL deterioration. A transient increase in gastrointestinal symptoms and a decrease in physical functioning were seen after neoadjuvant chemoradiation. In those patients who underwent surgical resection, most domains returned back to baseline levels by 6 months.
Collapse
Affiliation(s)
- Pablo E Serrano
- Department of Surgery, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Joseph M Herman
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kent A Griffith
- Center for Cancer Biostatistics, Biostatistics Unit, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Mark M Zalupski
- Center for Cancer Biostatistics, Biostatistics Unit, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Edward J Kim
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Tanios S Bekaii-Saab
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Edgar Ben-Josef
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Laura A Dawson
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Jolie Ringash
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada; Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Alice C Wei
- Department of Surgery, University Health Network, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada; Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
37
|
Blum D, Koeberle D, Omlin A, Walker J, Von Moos R, Mingrone W, deWolf-Linder S, Hayoz S, Kaasa S, Strasser F, Ribi K. Feasibility and acceptance of electronic monitoring of symptoms and syndromes using a handheld computer in patients with advanced cancer in daily oncology practice. Support Care Cancer 2014; 22:2425-34. [DOI: 10.1007/s00520-014-2201-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 03/04/2014] [Indexed: 10/25/2022]
|
38
|
Ghosn M, Kourie HR, Karak FE, Hanna C, Antoun J, Nasr D. Optimum chemotherapy in the management of metastatic pancreatic cancer. World J Gastroenterol 2014; 20:2352-2357. [PMID: 24605032 PMCID: PMC3942838 DOI: 10.3748/wjg.v20.i9.2352] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/24/2013] [Accepted: 01/15/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most devastating solid tumors, and it remains one of the most difficult to treat. The treatment of metastatic pancreatic cancer (MPC) is systemic, based on chemotherapy or best supportive care, depending on the performance status of the patient. Two chemotherapeutical regimens have produced substantial benefits in the treatment of MPC: gemcitabine in 1997; and FOLFIRIONOX in 2011. FOLFIRINOX improved the natural history of MPC, with overall survival (OS) of 11.1 mo. Nab-paclitaxel associated with gemcitabine is a newly approved regimen for MPC, with a median OS of 8.6 mo. Despite multiple trials, this targeted therapy was not efficient in the treatment of MPC. Many new molecules targeting the proliferation and survival pathways, immune response, oncofetal signaling and the epigenetic changes are currently undergoing phase I and II trials for the treatment of MPC, with many promising results.
Collapse
|
39
|
Solimando DA, Waddell JA. Capecitabine and Gemcitabine (CapGem, CG, GemCap) for Advanced Pancreatic and Biliary Tract Cancer. Hosp Pharm 2014; 49:127-33. [DOI: 10.1310/hpj4902-127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The complexity of cancer chemotherapy requires pharmacists be familiar with the complicated regimens and highly toxic agents used. This column reviews various issues related to preparation, dispensing, and administration of antineoplastic therapy, and the agents, both commercially available and investigational, used to treat malignant diseases.
Collapse
|
40
|
Gaertner J, Becker G. Patients with advanced disease: the value of patient reported outcomes. Oncol Res Treat 2014; 37:7-8. [PMID: 24613902 DOI: 10.1159/000358498] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 01/10/2014] [Indexed: 11/19/2022]
Affiliation(s)
- Jan Gaertner
- Department of Palliative Care, University Medical Center Freiburg, Freiburg i.Br., Germany
| | | |
Collapse
|
41
|
Bernhard J, Dietrich D, Glimelius B, Bodoky G, Scheithauer W, Herrmann R. Clinical benefit response in pancreatic cancer trials revisited. Oncol Res Treat 2014; 37:42-8. [PMID: 24613908 DOI: 10.1159/000357965] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/11/2013] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Clinical benefit response (CBR), based on changes in pain, Karnofsky performance status, and weight, is an established palliative endpoint in trials for advanced gastrointestinal cancer. We investigated whether CBR is associated with survival, and whether CBR reflects a wide-enough range of domains to adequately capture patients' perception. METHODS CBR was prospectively evaluated in an international phase III chemotherapy trial in patients with advanced pancreatic cancer (n = 311) in parallel with patient-reported outcomes (PROs). RESULTS The median time to treatment failure was 3.4 months (range: 0-6). The majority of the CBRs (n = 39) were noted in patients who received chemotherapy for at least 5 months. Patients with CBR (n = 62) had longer survival than non-responders (n = 182) (hazard ratio = 0.69; 95% confidence interval: 0.51-0.94; p = 0.013). CBR was predicted with a sensitivity and specificity of 77-80% by various combinations of 3 mainly physical PROs. A comparison between the duration of CBR (n = 62, median = 8 months, range = 4-31) and clinically meaningful improvements in the PROs (n = 100-116; medians = 9-11 months, range = 4-24) showed similar intervals. CONCLUSION CBR is associated with survival and mainly reflects physical domains. Within phase III chemotherapy trials for advanced gastrointestinal cancer, CBR can be replaced by a PRO evaluation, without losing substantial information but gaining complementary information.
Collapse
|
42
|
Li J, Podoltsev N, Saif MW. Management of advanced pancreatic cancer. Expert Rev Clin Pharmacol 2014; 2:527-41. [DOI: 10.1586/ecp.09.30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
43
|
Predictors of satisfaction with treatment decision, decision-making preferences, and main treatment goals in patients with advanced cancer. Support Care Cancer 2013; 21:3085-93. [PMID: 23828394 DOI: 10.1007/s00520-013-1886-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 06/19/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE This study investigated satisfaction with treatment decision (SWTD), decision-making preferences (DMP), and main treatment goals, as well as evaluated factors that predict SWTD, in patients receiving palliative cancer treatment at a Swiss oncology network. PATIENTS AND METHODS Patients receiving a new line of palliative treatment completed a questionnaire 4-6 weeks after the treatment decision. Patient questionnaires were used to collect data on sociodemographics, SWTD (primary outcome measure), main treatment goal, DMP, health locus of control (HLoC), and several quality of life (QoL) domains. Predictors of SWTD (6 = worst; 30 = best) were evaluated by uni- and multivariate regression models. RESULTS Of 480 participating patients in eight hospitals and two private practices, 445 completed all questions regarding the primary outcome measure. Forty-five percent of patients preferred shared, while 44 % preferred doctor-directed, decision-making. Median duration of consultation was 30 (range: 10-200) minutes. Overall, 73 % of patients reported high SWTD (≥24 points). In the univariate analyses, global and physical QoL, performance status, treatment goal, HLoC, prognosis, and duration of consultation were significant predictors of SWTD. In the multivariate analysis, the only significant predictor of SWTD was duration of consultation (p = 0.01). Most patients indicated hope for improvement (46 %), followed by hope for longer life (26 %) and better quality of life (23 %), as their main treatment goal. CONCLUSION Our results indicate that high SWTD can be achieved in most patients with a 30-min consultation. Determining the patient's main treatment goal and DMP adds important information that should be considered before discussing a new line of palliative treatment.
Collapse
|
44
|
Metastatic pancreatic cancer: are we making progress in treatment? Gastroenterol Res Pract 2012; 2012:898931. [PMID: 23304129 PMCID: PMC3523135 DOI: 10.1155/2012/898931] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Accepted: 11/02/2012] [Indexed: 12/12/2022] Open
Abstract
Development of systemic treatment for advanced pancreatic cancer (APC) has been challenging. After fluorouracil, gemcitabine (GEM) became the treatment of choice based on its benefit of symptom relief. Many cytotoxic agents have been combined with GEM in search of regimens with improved survival benefit. However, there were only marginal benefits in people with good performance status. Recently, the combination regimen consisting of oxaliplatin, irinotecan, fluorouracil, and leucovorin (FOLFIRINOX) was found to achieve unprecedented survival benefit and has become the preferred option for patients with good clinical conditions. On the other hand, many biological agents have been combined with GEM, but only erlotinib was found to derive statistically significant survival advantage. However, the effect was too small to be appreciated clinically. The effort in development of targeted therapy in APC continues. This paper summarized key findings in the development of chemotherapy and targeted therapy for APC patients and discussed future directions in management.
Collapse
|
45
|
Sun C, Ansari D, Andersson R, Wu DQ. Does gemcitabine-based combination therapy improve the prognosis of unresectable pancreatic cancer? World J Gastroenterol 2012; 18:4944-4958. [PMID: 23002368 PMCID: PMC3447278 DOI: 10.3748/wjg.v18.i35.4944] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 02/01/2012] [Accepted: 04/12/2012] [Indexed: 02/06/2023] Open
Abstract
AIM To assess whether gemcitabine-based combination therapy improves the prognosis of unresectable pancreatic cancer compared with gemcitabine treatment alone. METHODS A quantitative up-to-date meta-analysis was undertaken to investigate the efficacy of gemcitabine-based combination treatment compared with gemcitabine monotherapy in locally advanced or metastatic pancreatic cancer. Inclusion was limited to high-quality randomized clinical trials. RESULTS Twenty-six studies were included in the present analysis, with a total of 8808 patients recruited. The studies were divided into four subgroups based on the different kinds of cytotoxic agents, including platinum, fluoropyrimidine, camptothecin and targeted agents. Patients treated with gemcitabine monotherapy had significantly lower objective response rate [risk ratio (RR), 0.72; 95% confidence interval (CI): 0.63-0.83; P < 0.001], and lower 1-year overall survival (RR, 0.90; 95%CI: 0.82-0.99; P = 0.04). Gemcitabine monotherapy caused fewer complications, including fewer grade 3-4 toxicities: including vomiting (RR, 0.75; 95%CI: 0.62-0.89; P = 0.001), diarrhea (RR, 0.66; 95%CI: 0.49-0.89; P = 0.006), neutropenia (RR, 0.88; 95%CI: 0.72-1.06; P = 0.18), anemia (RR, 0.96; 95%CI: 0.82-1.12; P = 0.60), and thrombocytopenia (RR, 0.76; 95%CI: 0.60-0.97; P = 0.03) compared with gemcitabine combination therapies. CONCLUSION Gemcitabine combination therapy provides a modest improvement of survival, but is associated with more toxicity compared with gemcitabine monotherapy.
Collapse
|
46
|
Zabernigg A, Giesinger JM, Pall G, Gamper EM, Gattringer K, Wintner LM, Sztankay MJ, Holzner B. Quality of life across chemotherapy lines in patients with cancers of the pancreas and biliary tract. BMC Cancer 2012; 12:390. [PMID: 22950826 PMCID: PMC3488526 DOI: 10.1186/1471-2407-12-390] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 08/31/2012] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND In patients with cancers of the pancreatic and biliary tract quality of life (QOL) improvement is the main treatment goal, since survival can be prolonged only marginally. Up to date, knowledge on QOL impairments throughout the entire treatment process, often including several chemotherapy lines, is scarce. Our study aimed at investigating QOL trajectories from adjuvant treatment to palliative 3rd-line therapy METHODS Patients were included in routine electronic patient-reported outcome monitoring at Kufstein County Hospital at the time of diagnosis and assessed with the EORTC QLQ-C30 during each chemotherapy cycle. RESULTS Eighty out of 147 patients with pancreatic cancer or cancer of the bile ducts treated at the Kufstein County Hospital, fulfilled inclusion criteria and could be included in the study (mean age 67.4 years; 53.8% women). Physical, Emotional and Cognitive Functioning, and Global QOL deteriorated across chemotherapy lines, whereas Fatigue, Pain, Dyspnoea, Sleeping Disturbances, Diarrhoea, and Taste Alterations increased. With regard to Physical Functioning, Global QOL, Fatigue, Dyspnoea, Diarrhoea and Taste Alterations, the patients receiving adjuvant or 1st-line palliative chemotherapy did not differ significantly. Most patients in 2nd- or 3rd-line chemotherapy showed significantly higher impairments and symptom burden. However, patients under 1st and 2nd-line treatment showed stable QOL trajectories, whereas 3rd-line patients perceived substantial deteriorations. CONCLUSIONS The results suggest early palliative treatment initiation to stabilise QOL on a level as high as possible. The continuous QOL improvement during adjuvant treatment, probably reflecting post-operative recovery, may indicate that deleterious effects of adjuvant chemotherapy on QOL are highly unlikely.
Collapse
Affiliation(s)
- August Zabernigg
- Department of Internal Medicine, Kufstein County Hospital, Endach 27, A-6330, Kufstein, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Involvement of p53 in gemcitabine mediated cytotoxicity and radiosensitivity in breast cancer cell lines. Gene 2012; 498:300-7. [DOI: 10.1016/j.gene.2012.01.099] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 01/30/2012] [Accepted: 01/30/2012] [Indexed: 11/21/2022]
|
48
|
Sequential FOLFOX-6 and gemcitabine for locally advanced and/or metastatic pancreatic cancer. Med Oncol 2012; 29:2831-7. [PMID: 22392197 DOI: 10.1007/s12032-012-0197-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 02/18/2012] [Indexed: 01/05/2023]
Abstract
This prospective phase II trial aims to evaluate the sequential FOLFOX-6 and gemcitabine followed by adapted maintenance for advanced pancreatic cancer. Treatment included FOLFOX-6 for 4 cycles, followed sequentially by gemcitabine for 3 cycles. Patients, who show clinical benefit after both sequences, will receive maintenance treatment based on the investigator's discretion. From January 2005 to June 2008, 32 patients with median age of 63 were included; 75% of patients had metastatic disease, 81% had pure adenocarcinoma, while 19% had adenocarcinoma with a neuroendocrine component. There were 22% PR and 22% SD resulting in 44% tumor growth control. Under FOLFOX, grade 3/4 toxicities were neutropenia in 8 patients, thrombocytopenia and anemia in 3 patients each, and diarrhea in 2 patients. Under Gem, grade 3/4 neutropenia was observed in 4 patients, thrombocytopenia and anemia were observed in 2 patients, and hand-foot syndrome was observed in 3 patients. The median TTP and OS were 4 and 10 months, respectively. In APC, FOLFOX-6 regimen followed by gemcitabine achieved an interesting RR within a tolerable level of toxicity. This regimen seems to warrant further investigation to confirm its efficacy.
Collapse
|
49
|
Ying JE, Zhu LM, Liu BX. Developments in metastatic pancreatic cancer: is gemcitabine still the standard? World J Gastroenterol 2012; 18:736-45. [PMID: 22371633 PMCID: PMC3286136 DOI: 10.3748/wjg.v18.i8.736] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Revised: 08/26/2011] [Accepted: 09/03/2011] [Indexed: 02/06/2023] Open
Abstract
In the past 15 years, we have seen few therapeutic advances for patients with pancreatic cancer, which is the fourth leading cause of cancer-related death in the United States. Currently, only about 6% of patients with advanced disease respond to standard gemcitabine therapy, and median survival is only about 6 mo. Moreover, phase III trials have shown that adding various cytotoxic and targeted chemotherapeutic agents to gemcitabine has failed to improve overall survival, except in cases in which gemcitabine combined with erlotinib show minimal survival benefit. Several meta-analyses have shown that the combination of gemcitabine with either a platinum analog or capecitabine may lead to clinically relevant survival prolongation, especially for patients with good performance status. Meanwhile, many studies have focused on the pharmacokinetic modulation of gemcitabine by fixed-dose administration, and metabolic or transport enzymes related to the response and toxicity of gemcitabine. Strikingly, a phase III trial in 2010 showed that, in comparison to gemcitabine alone, the FOLFIRINOX regimen in patients with advanced disease and good performance status, produced better median overall survival, median progression-free survival, and objective response rates. This regimen also resulted in greater, albeit manageable toxicity.
Collapse
|
50
|
Lee S, Kim K, Jung H, Lee S, Cheon S, Kim S, Eo W, Choi W. Efficacy and safety of standardized allergen-removed Rhus verniciflua Stokes extract in patients with advanced or metastatic pancreatic cancer: a Korean single-center experience. Oncology 2011; 81:312-8. [PMID: 22179506 DOI: 10.1159/000334695] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 10/22/2011] [Indexed: 01/05/2023]
Abstract
BACKGROUND Pancreatic cancer has the worst prognosis because of poor response to conventional therapy. We investigated the clinical feasibility of the standardized allergen-removed Rhus verniciflua Stokes (aRVS) extract as a potential therapeutic agent for advanced or metastatic pancreatic cancer. PATIENTS AND METHODS From July 2006 to June 2010, patients with advanced or metastatic pancreatic adenocarcinoma were checked in our institution. After applying inclusion/exclusion criteria, 42 patients were eligible for the final analysis. Overall survival, clinical benefit and adverse events of these patients treated with aRVS in the aftercare period were determined. RESULTS In May 2011, 39 patients had died and the remaining 3 patients were alive with evidence of disease. The mean RVS administration period was 3.86 months (95% confidence interval 2.52-5.20). The median overall survival for the entire population was 7.87 months (95% confidence interval 5.14-10.59), and the 1-year survival rate was 26.2%, which is compatible with external controls. Using univariate and multivariate analyses, aRVS treatment including performance status and prognostic index significantly affected overall survival. A clinical benefit response was also shown by aRVS treatment which was not dependent on concurrent chemotherapy. Adverse reactions to aRVS treatment were mostly mild and self-limiting. CONCLUSIONS The standardized aRVS extract might be beneficial for patients with advanced or metastatic pancreatic cancer since it positively affected overall survival and clinical symptoms without significant adverse effects.
Collapse
Affiliation(s)
- Sanghun Lee
- Department of Clinical Oncology, Integrative Cancer Center, Kyunghee University Hospital at Gangdong, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|