1
|
Wainberg ZA, M O'Reilly E. NALIRIFOX in the frontline for metastatic pancreatic cancer: evidence beyond NAPOLI 3. Nat Rev Clin Oncol 2024:10.1038/s41571-024-00952-5. [PMID: 39455872 DOI: 10.1038/s41571-024-00952-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Affiliation(s)
- Zev A Wainberg
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Eileen M O'Reilly
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
2
|
Netto D, Frizziero M, Foy V, McNamara MG, Backen A, Hubner RA. Systemic Therapy for Metastatic Pancreatic Cancer-Current Landscape and Future Directions. Curr Oncol 2024; 31:5206-5223. [PMID: 39330013 PMCID: PMC11430697 DOI: 10.3390/curroncol31090385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a significant cause of cancer-associated mortality, with a rising global incidence. A paucity of strong predictive risk factors mean screening programmes are difficult to implement. Historically, a lack of identifiable and actionable driver mutations, coupled with a relatively immunosuppressed tumour microenvironment, has led to a reliance on cytotoxic chemotherapy. The NAPOLI-3 trial has reported data supporting consideration of NALIRIFOX as a new first-line standard of care. Kirsten Rat Sarcoma Virus (KRAS) G12D mutations are present in >90% of all PDAC's; exciting breakthroughs in small molecule inhibitors targeting KRAS G12D may open new modalities of treatment, and therapies targeting multiple KRAS mutations are also in early clinical trials. Although immunotherapy strategies to date have been disappointing, combination with chemotherapy and/or small molecule inhibitors hold promise and warrant further exploration.
Collapse
Affiliation(s)
- Daniel Netto
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Melissa Frizziero
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Victoria Foy
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Mairéad G McNamara
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Alison Backen
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Richard A Hubner
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| |
Collapse
|
3
|
Ueno H, Shimizu K, Fukui A, Nii M, Koto R, Unno M. Evaluation of the Treatment Duration of Japanese Patients With Pancreatic Cancer in a Real-World Setting Using a Large Hospital Claims Database: The SUISEI Study. Pancreas 2024; 53:e492-e500. [PMID: 38767967 DOI: 10.1097/mpa.0000000000002321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
OBJECTIVES To clarify the treatment reality of pancreatic cancer in Japan, focusing on treatment duration and time to death. MATERIALS AND METHODS We retrospectively analyzed Japanese hospital claims data for patients diagnosed with pancreatic cancer between April 2009 and October 2018 to investigate treatment patterns, duration of first-line chemotherapy, and time to death. RESULTS Of 81,185 eligible patients, 54.2% were male, the mean age was 71.7 years, and 23.3% (n = 18,884) received chemotherapy as primary treatment. The median treatment duration was 14.1 weeks for the 6.7% of patients who received oxaliplatin, irinotecan, fluorouracil, and leucovorin (FOLFIRINOX; recommended first-line regimen) and 16.9 weeks for the 30.2% of patients who received gemcitabine plus nab-paclitaxel (GEM + nab-PTX). Time to death for patients who received FOLFIRINOX or GEM + nab-PTX was similar (15.4 and 14.8 months, respectively). The duration of first-line chemotherapy regimens tended to increase annually for both regimens. The time to death for all first-line chemotherapy regimens also increased annually. CONCLUSIONS This study revealed the treatment reality of pancreatic cancer in the real-world Japanese setting. Treatment duration and time to death tended to increase over time and did not differ numerically between FOLFIRINOX and GEM + nab-PTX.
Collapse
Affiliation(s)
- Hideki Ueno
- From the Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo
| | - Kyoko Shimizu
- Department of Gastroenterology, Tokyo Women's Medical University, Tokyo
| | | | | | | | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
4
|
Balar PC, Apostolopoulos V, Chavda VP. A new era of immune therapeutics for pancreatic cancer: Monoclonal antibodies paving the way. Eur J Pharmacol 2024; 969:176451. [PMID: 38408598 DOI: 10.1016/j.ejphar.2024.176451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/06/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
Pancreatic cancer, particularly pancreatic ductal adenocarcinoma, remains a devastating disease with a dismal prognosis and limited survival rates. Despite various drug treatments and regimens showing promise in managing the disease, the clinical outcomes have not significantly improved. Immunotherapy however, has become a forefront area in pancreatic cancer treatment. This approach comprises a range of agents, including small molecule drugs, antibodies, combination therapies, and vaccines. In the last 5-8 years, there has been an upsurge of research into the use of monoclonal antibodies to block receptors on cancer or immune cells, revolutionising cancer treatment and management. Several targets have been identified and studied, with the most encouraging noted in relation to checkpoint markers, namely, antibodies targeting anti-programmed cell death 1 (PD-1) and its receptor PD-L1. Herein, we present the clinical developments in immunotherapy in the last 5 years especially those which have been tested in humans against pancreatic cancer.
Collapse
Affiliation(s)
- Pankti C Balar
- Pharmacy Section, L.M. College of Pharmacy, Ahmedabad, India
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Werribee Campus, Melbourne, VIC, 3030, Australia
| | - Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, India.
| |
Collapse
|
5
|
Piątek M, Bieńkowski M, Kuśnierz K, Pilch-Kowalczyk J, Imielska-Zdunek D, Mrowiec S, Lampe P, Radecka B, Nawrocki S. Combination of modified FOLFIRINOX with stereotactic body radiotherapy as an induction therapy for locally advanced pancreatic adenocarcinoma - a prospective single-arm study. Contemp Oncol (Pozn) 2024; 28:15-30. [PMID: 38800534 PMCID: PMC11117165 DOI: 10.5114/wo.2024.137760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/14/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Radical resection is the only potentially curative treatment for pancreatic adenocarcinoma; however, only a minor fraction of patients are eligible for resection. Induction therapy may be offered to patients, but the response rate in cases with significant vascular involvement is limited. This study aimed to evaluate the efficacy and safety of modified of FOLFIRINOX chemotherapy (mFFX) + stereotactic body radiotherapy (SBRT) in combination as induction therapy for locally advanced pancreatic carcinoma. The primary endpoints were the resection rate and one-year overall survival (OS). The secondary endpoints were progression-free survival (PFS), toxicity, and quality of live (QoL). Material and methods Thirty patients with locally advanced pancreatic adenocarcinoma were treated with 6 cycles of mFFX, followed by SBRT and additional 3 cycles of mFFX. The response was measured prior to SBRT and after regimen completion. In the absence of disease progression, the patients were referred for surgery. The patients were requested to complete quality of life questionnaires (QLQ)-C30 and QLQ-PAN26 questionnaires biweekly. Results On the first evaluation, disease control was noted in 26 (86.7%) patients. Stereotactic body radiotherapy was performed in 20 patients. Twelve patients underwent laparotomy, with radical resection possible in 3 cases. The one-year OS rate was 63.3%. Overall, 11 grade ≥ 3 adverse events were noted. No deterioration in the overall QoL was observed. The median PFS was 7.53 months. Conclusions The expected resection rate of ≥ 30% was not achieved. However, the combination was associated with good local control, low adverse event rate, and good QoL, which advocate its further investigation in this clinical situation.
Collapse
Affiliation(s)
- Michał Piątek
- Department of Oncology, Medical University of Silesia, Katowice, Poland
- Department of Oncology, Institute of Medical Sciences, University of Opole, Opole, Poland
| | - Michał Bieńkowski
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Poland
| | - Katarzyna Kuśnierz
- Department of Gastrointestinal Surgery, Medical University of Silesia, Katowice, Poland
| | - Joanna Pilch-Kowalczyk
- Department of Radiology and Nuclear Medicine, Medical University of Silesia, Katowice, Poland
| | - Dorota Imielska-Zdunek
- Radiotherapy Division, prof. K. Gibiński Memorial University Clinical Centre, Silesian Medical University, Katowice, Poland
| | - Sławomir Mrowiec
- Department of Gastrointestinal Surgery, Medical University of Silesia, Katowice, Poland
| | - Paweł Lampe
- Department of Gastrointestinal Surgery, Medical University of Silesia, Katowice, Poland
| | - Barbara Radecka
- Department of Oncology, Institute of Medical Sciences, University of Opole, Opole, Poland
| | - Sergiusz Nawrocki
- Department of Radiotherapy, Medical University of Silesia, Katowice, Poland
- Oncology Department, Faculty of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
6
|
Yuen JG, Hwang GR, Fesler A, Intriago E, Pal A, Ojha A, Ju J. Development of gemcitabine-modified miRNA mimics as cancer therapeutics for pancreatic ductal adenocarcinoma. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200769. [PMID: 38596306 PMCID: PMC10869788 DOI: 10.1016/j.omton.2024.200769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/23/2023] [Accepted: 01/19/2024] [Indexed: 04/11/2024]
Abstract
Despite the recent advancement in diagnosis and therapy, pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is still the most lethal cancer with a low five-year survival rate. There is an urgent need to develop new therapies to address this issue. In this study, we developed a treatment strategy by modifying tumor suppressor miRNAs, miR-15a and miR-194, with the chemotherapeutic gemcitabine (Gem) to create Gem-modified mimics, Gem-miR-15a and Gem-miR-194, respectively. In a panel of PDAC cell lines, we found that Gem-miR-15a and Gem-miR-194 induce cell-cycle arrest and apoptosis, and these mimics are potent inhibitors with IC50 values up to several hundred fold less than their native counterparts or Gem alone. Furthermore, we found that Gem-miR-15a and Gem-miR-194 retained miRNA function by downregulating the expression of several key targets including WEE1, CHK1, BMI1, and YAP1 for Gem-miR-15a, and FOXA1 for Gem-miR-194. We also found that our Gem-modified miRNA mimics exhibit an enhanced efficacy compared to Gem in patient-derived PDAC organoids. Furthermore, we observed that Gem-miR-15a significantly inhibits PDAC tumor growth in vivo without observing any noticeable signs of toxicity. Overall, our results demonstrate the therapeutic potential of Gem-modified miRNAs as a treatment strategy for PDAC.
Collapse
Affiliation(s)
- John G. Yuen
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Medical Scientist Training Program, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Ga-Ram Hwang
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | | | - Erick Intriago
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Amartya Pal
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Anushka Ojha
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jingfang Ju
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- The Northport Veteran’s Administration Medical Center, Northport, NY 11768, USA
| |
Collapse
|
7
|
Gueiderikh A, Tarabay A, Abdelouahab M, Smolenschi C, Tanguy ML, Valery M, Malka D, Pudlarz T, Fuerea A, Boige V, Hollebecque A, Ducreux M, Boilève A. Pancreatic adenocarcinoma third line systemic treatments: a retrospective cohort study. BMC Cancer 2024; 24:272. [PMID: 38408958 PMCID: PMC10898186 DOI: 10.1186/s12885-024-12016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/16/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Chemotherapy for metastatic pancreatic adenocarcinoma (PDAC) primarily relies on FOLFIRINOX (LV5FU- irinotecan - Oxaliplatine) and Gemcitabine - Nab-Paclitaxel in the first-line setting. However, second-lines remain less well-defined and there is limited data regarding third-line treatments. The objective of our study was to determine the proportion of patients advancing to third line chemotherapy, to outline the various third-line chemotherapy regimens used in routine practice and to evaluate their respective efficacy. METHODS A retrospective single-center cohort from 2010-2022 compiled baseline characteristics, treatment outcomes and survival of PDAC patients who received at least one chemotherapy line in a French tertiary-center. Overall survivals (OS) were analyzed using a Cox multivariable model. RESULTS In total, 676 patients were included, with a median follow-up time of 69.4 months, (Interquartile Range (IQR) = 72.1). Of these, 251 patients (37%) that proceeded to 3rd-line chemotherapy. The median PFS in 3rd line was 2.03 months, [CI95%: 1.83, 2.36]. The median 3rd line overall survival was 5.5 months, [CI95%: 4.8, 6.3]. In multivariable analysis erlotinib-based chemotherapy was found to be deleterious (HR=2.38, [CI95%: 1.30, 4.34], p=0.005) compared to fluoropyrimidine-based chemotherapy in terms of 3rd line overall survival while gemcitabine monotherapy showed a tendency towards negative outcomes. First and 2nd line chemotherapies sequence didn't influence 3rd line outcome. CONCLUSION In our cohort, one-third of treated patients proceeded to 3rd line chemotherapy resulting in a 5.5 months median 3rd line OS, consistent with treatments at advanced stage. Our results argue against the use of erlotinib and gemcitabine monotherapy.
Collapse
Affiliation(s)
- A Gueiderikh
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
- Université Paris Saclay, 91471, Orsay, France
| | - A Tarabay
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
| | - M Abdelouahab
- Département de statistiques, Gustave Roussy, 94800, Villejuif, France
| | - C Smolenschi
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
- Gustave Roussy, DITEP, 94800, Villejuif, France
| | - M L Tanguy
- Département de statistiques, Gustave Roussy, 94800, Villejuif, France
| | - M Valery
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
| | - D Malka
- Département d'oncologie médicale, Institut Mutualiste Montsouris, 75014, Paris, France
| | - T Pudlarz
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
| | - A Fuerea
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
| | - V Boige
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
| | - A Hollebecque
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
- Gustave Roussy, DITEP, 94800, Villejuif, France
| | - M Ducreux
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France
- Université Paris Saclay, 91471, Orsay, France
| | - A Boilève
- Département de médecine oncologique, Gustave Roussy, 94800, Villejuif, France.
- Université Paris Saclay, 91471, Orsay, France.
| |
Collapse
|
8
|
Besselink MG, Wilmink JW. Treating metastatic pancreatic ductal adenocarcinoma: NALIRIFOX as new standard? Lancet 2023; 402:1217-1218. [PMID: 37708905 DOI: 10.1016/s0140-6736(23)01521-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 09/16/2023]
Affiliation(s)
- Marc G Besselink
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam 1081 HV, Netherlands.
| | - Johanna W Wilmink
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam 1081 HV, Netherlands
| |
Collapse
|
9
|
Singhal R, Rogers SC, Lee JH, Ramnaraign B, Sahin I, Fabregas JC, Thomas RM, Hughes SJ, Nassour I, Hitchcock K, Russell K, Kayaleh O, Turk A, Zlotecki R, DeRemer DL, George TJ. A phase II study of neoadjuvant liposomal irinotecan with 5-FU and oxaliplatin (NALIRIFOX) in pancreatic adenocarcinoma. Future Oncol 2023; 19:1841-1851. [PMID: 37753702 PMCID: PMC10594143 DOI: 10.2217/fon-2023-0256] [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: 03/28/2023] [Accepted: 08/01/2023] [Indexed: 09/28/2023] Open
Abstract
For patients with localized pancreatic cancer with minimal vascular involvement, optimal survivability requires a multidisciplinary approach of surgical resection and systemic chemotherapy. FOLFIRINOX is a combination chemotherapy regimen that offers promising results in the perioperative and metastatic settings; however, it can cause significant adverse effects. Such toxicity can negatively impact some patients, resulting in chemotherapy discontinuation or surgical unsuitability. In an effort to reduce toxicities and optimize outcomes, this investigation explores the safety and feasibility of substituting liposomal irinotecan (nal-IRI) for nonliposomal irinotecan to improve tumor drug delivery and potentially reduce toxicity. This regimen, NALIRIFOX, has the potential to be both safer and more effective when administered in the preoperative setting.
Collapse
Affiliation(s)
- Ruchi Singhal
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Sherise C Rogers
- Department of Medicine, Division of Hematology & Oncology, University of Florida, Gainesville, FL, USA
| | - Ji-Hyun Lee
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Brian Ramnaraign
- Department of Medicine, Division of Hematology & Oncology, University of Florida, Gainesville, FL, USA
| | - Ilyas Sahin
- Department of Medicine, Division of Hematology & Oncology, University of Florida, Gainesville, FL, USA
| | - Jesus C Fabregas
- Memorial Cancer Institute, Florida Atlantic University, Hollywood, FL, USA
| | - Ryan M. Thomas
- Department of Surgery, University of Florida, Gainesville, FL, USA
| | - Steven J Hughes
- Department of Surgery, University of Florida, Gainesville, FL, USA
| | - Ibrahim Nassour
- Department of Surgery, University of Florida, Gainesville, FL, USA
| | | | | | - Omar Kayaleh
- Orlando Health Cancer Institute, Orlando, FL, USA
| | - Anita Turk
- Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Robert Zlotecki
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | - David L DeRemer
- College of Pharmacy, University of Florida, Department of Pharmacotherapy & Translational Research, Gainesville, FL, USA
| | - Thomas J George
- Department of Medicine, Division of Hematology & Oncology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
10
|
Yuen JG, Hwang GR, Fesler A, Intriago E, Pal A, Ojha A, Ju J. Development of Gemcitabine-Modified miRNA Mimics as Cancer Therapeutics for Pancreatic Ductal Adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553255. [PMID: 37645827 PMCID: PMC10462072 DOI: 10.1101/2023.08.14.553255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Pancreatic cancer, including its most common subtype, pancreatic adenocarcinoma (PDAC), has the lowest five-year survival rate among patients with pancreatic cancer in the United States. Despite advancements in anticancer treatment, the overall median survival for patients with PDAC has not dramatically improved. Therefore, there is an urgent need to develop new strategies of treatment to address this issue. Non-coding RNAs, including microRNAs (miRNAs), have been found to have major roles in carcinogenesis and the subsequent treatment of various cancer types like PDAC. In this study, we developed a treatment strategy by modifying tumor suppressor miRNAs, hsa-miRNA-15a (miR-15a) and hsa-miRNA-194-1 (miR-194), with the nucleoside analog chemotherapeutic gemcitabine (Gem) to create Gem-modified mimics of miR-15a (Gem-miR-15a) and miR-194 (Gem-miR-194). In a panel of PDAC cell lines, we found that Gem-miR-15a and Gem-miR-194 induce cell cycle arrest and apoptosis, and these mimics are potent inhibitors with IC 50 values up to several hundred fold less than their native counterparts or Gem alone. Furthermore, we found that Gem-miR-15a and Gem-miR-194 retained miRNA function by downregulating the expression of several key targets including WEE1, CHK1, BMI1, and YAP1 for Gem-miR-15a, and FOXA1 for Gem-miR-194. We also found that our Gem-modified miRNA mimics exhibit an enhanced efficacy compared to Gem alone in patient-derived PDAC organoids. Furthermore, we observed that Gem-miR-15a significantly inhibits PDAC tumor growth in vivo without observing any noticeable signs of toxicity. Overall, our results demonstrate the therapeutic potential of Gem-modified miRNAs as a treatment strategy for PDAC. One Sentence Summary Yuen and Hwang et. al. have developed a potent therapeutic strategy for patients with pancreatic cancer by modifying microRNAs with gemcitabine.
Collapse
|
11
|
Jung K, Choi S, Song H, Kwak K, Anh S, Jung JH, Kim B, Ahn J, Kim J, Hwang JH, Lee JC. Real-world dose reduction of standard and modified FOLFIRINOX in metastatic pancreatic cancer: a systematic review, evidence-mapping, and meta-analysis. Ther Adv Med Oncol 2023; 15:17588359231175441. [PMID: 37441327 PMCID: PMC10333643 DOI: 10.1177/17588359231175441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 04/25/2023] [Indexed: 07/15/2023] Open
Abstract
Background FOLFIRINOX, used in metastatic pancreatic cancer (MPC), is highly efficacious but also toxic. Various dose modifications for FOLFIRINOX have been introduced to reduce toxicity. However, these studies lack a unified pattern for 'planned' dose modification, and the 'actually administered' dose varied more. Objective To map a 10-year trend for 'planned' and 'actual' doses of FOLFIRINOX and investigate the clinical outcomes according to dose modification. Data sources and methods A comprehensive systematic literature search was conducted from January 2011 to September 2021. All studies for FOLFIRINOX as first-line treatment in MPC were considered. Selected studies were firstly classified according to prospective versus retrospective research, secondly standard versus modified FOLFIRINOX, and thirdly 'planned' versus 'actual' dose. For evidence-mapping for the trend of dose modification, we developed a web-based interactive bubble-plot program (www.RDI-map.com). Objective response rate (ORR) and hematologic toxicity were set as endpoints for the comparison of clinical outcomes according to dose modification. Results A total of 37 studies were identified for evidence-mapping (11 prospective and 26 retrospective studies). There were 12 different types of 'planned' dose modification in FOLFIRINOX ranging 75-100% oxaliplatin, 75-100% irinotecan, 0-100% 5-fluorouracil (5-FU) bolus, and 75-133% 5-FU continuous injection. The 'actual' dose further decreased to 54-96%, 61-88%, 0-92%, and 63-98%, respectively. For the standard versus modified FOLFIRINOX, the ORR was 28.2% (95% CI: 22.5-33.9%) and 33.8% (95% CI: 30.3-37.3%), respectively (p = 0.100), and the incidence of febrile neutropenia was 11.6% (95% CI: 0-16.0%) and 5.5% (95% CI: 0-8.9%), respectively (p = 0.030). Conclusions RDI-map.com enables multifactorial evidence-mapping for practical FOLFIRINOX dose reduction. The pattern of dose modification was not consistent across studies, and there was a significant gap between the 'planned' and 'actual' doses. Modified FOLFIRINOX showed similar efficacy to the standard regimen with reduced incidence of febrile neutropenia.
Collapse
Affiliation(s)
| | | | - Hyunjoo Song
- School of Computer Science and Engineering,
Soongsil University, Seoul, Korea
| | - Kyuhan Kwak
- School of Computer Science and Engineering,
Soongsil University, Seoul, Korea
| | - Soyeon Anh
- Division of Statistics, Medical Research
Collaborating Center, Seoul National University Bundang Hospital, Seongnam,
Korea
| | - Jae Hyup Jung
- Department of Internal Medicine, Seoul National
University Bundang Hospital, Seongnam, Korea
| | - Bomi Kim
- Department of Internal Medicine, Seoul National
University Bundang Hospital, Seongnam, Korea
| | - Jinwoo Ahn
- Department of Internal Medicine, Seoul National
University Bundang Hospital, Seongnam, Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National
University Bundang Hospital, Seongnam, Korea
- College of Medicine, Seoul National
University, Seoul, Korea
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul
National University Bundang Hospital, Seongnam, Korea
- College of Medicine, Seoul National
University, Seoul, Korea
| | | |
Collapse
|
12
|
Maman Y, Goykhman Y, Yakir O, Barenboim A, Geva R, Peles-Avraham S, Wolf I, Klausner JM, Lahat G, Lubezky N. Adjuvant FOLFIRINOX in Patients with Resectable Pancreatic Cancer Is Effective but Rarely Feasible in Real Life: Is Neoadjuvant FOLFIRINOX a Better Option? Cancers (Basel) 2023; 15:cancers15113049. [PMID: 37297011 DOI: 10.3390/cancers15113049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND The recommended treatment for resectable pancreatic cancer (PC) is resection followed by adjuvant FOLFIRINOX. We assessed the proportion of patients that managed to complete the 12 courses of adjuvant FOLFIRINOX and compared their outcome with that of patients with borderline resectable pancreatic cancer (BRPC) who underwent resection after neoadjuvant FOLFIRINOX. METHODS A retrospective analysis was performed on a prospectively maintained database of all PC patients who underwent resection with (2/2015-12/2021) or without (1/2018-12/2021) neoadjuvant therapy. RESULTS A total of 100 patients underwent upfront resection, and 51 patients with BRPC received neoadjuvant treatment. Only 46 resection patients started adjuvant FOLFIRINOX, and only 23 completed 12 courses. The main reasons for not starting/completing adjuvant therapy were poor tolerance and rapid recurrence. Significantly more patients in the neoadjuvant group received at least six FOLFIRINOX courses (80.4% vs. 31%, p < 0.001). Patients who completed at least 6 courses, either pre- or postoperatively, had better overall survival (p = 0.025) than those who did not. In spite of having more advanced disease, the neoadjuvant group had comparable overall survival (p = 0.062) regardless of the number of treatment courses. CONCLUSION Only a minority of patients (23%) undergoing upfront pancreatic resection completed the planned 12 courses of FOLFIRINOX. Patients who received neoadjuvant treatment were significantly more likely to receive at least six treatment courses. Patients receiving at least six courses had better overall survival than those who received fewer than six courses, regardless of the timing of treatment relative to surgery. Potential ways to increase chemotherapy adherence, such as administering treatment before surgery, should be considered.
Collapse
Affiliation(s)
- Yossi Maman
- Departments of Surgery, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Yaacov Goykhman
- Departments of HPB and Transplant Surgery, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Oz Yakir
- Departments of HPB and Transplant Surgery, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Alex Barenboim
- Departments of Surgery, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Ravit Geva
- Institute of Oncology, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Sharon Peles-Avraham
- Institute of Oncology, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Ido Wolf
- Institute of Oncology, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Joseph M Klausner
- Departments of Surgery, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Guy Lahat
- Departments of Surgery, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Nir Lubezky
- Departments of HPB and Transplant Surgery, Tel-Aviv Medical Center, Sackler School of Medicine, The Nicholas and Elizabeth Cathedra of Experimental Surgery, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
13
|
Sarfraz H, Saha A, Jhaveri K, Kim DW. Review of Current Systemic Therapy and Novel Systemic Therapy for Pancreatic Ductal Adenocarcinoma. Curr Oncol 2023; 30:5322-5336. [PMID: 37366887 PMCID: PMC10296812 DOI: 10.3390/curroncol30060404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/21/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND This review aims to describe the systemic treatment options for pancreatic ductal adenocarcinoma and includes a summary of the current treatments as well as the ongoing clinical trials which may be efficacious in the treatment of this aggressive malignancy. METHODS A literature review was performed using MEDLINE/PubMed between August 1996 and February 2023. The reviewed studies are categorized into these categories: current standard of care treatments, targeted therapies, immunotherapy and clinical trials. The current treatment modality for the treatment of advanced pancreatic cancer is mainly systemic chemotherapy. RESULTS The introduction of polychemotherapy regimens including gemcitabine/nab-paclitaxel and FOLFIRINOX (oxaliplatin, irinotecan, folinic acid and fluorouracil) has improved the clinical outcome of advanced pancreatic cancer. For further improvement in clinical outcomes, several novel approaches have been extensively studied in pancreatic cancer. The review discusses the current standard chemotherapy regimen and the novel treatment options in the field. CONCLUSIONS While there are novel treatments being explored for metastatic pancreatic, it remains a debilitating and aggressive disease with high mortality that warrants continued efforts to advance therapeutic options.
Collapse
Affiliation(s)
| | | | | | - Dae Won Kim
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (H.S.); (A.S.); (K.J.)
| |
Collapse
|
14
|
Carvalho de Brito AB, Riechelmann RP, Fonseca de Jesus VH. Impact of Granulocyte Colony-Stimulating Factor (G-CSF) on the Outcomes of Patients With Metastatic Pancreatic Adenocarcinoma (MPA) During First-Line Treatment With FOLFIRINOX: A Single-Center Retrospective Analysis. Cancer Control 2023; 30:10732748221149543. [PMID: 36592369 PMCID: PMC9829887 DOI: 10.1177/10732748221149543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION The role of primary prophylaxis (PP) with granulocyte colony-stimulating factor (G-CSF) for patients with metastatic pancreatic adenocarcinoma (MPA) treated with FOLFIRINOX is unknown. We aimed to compare the frequencies of grades 3 or 4 neutropenia (G3/4N) and febrile neutropenia (FN) and survival outcomes according to the use of PP. METHODS This is a retrospective study. We included patients with pathologically confirmed MPA treated with FOLFIRINOX in first-line. Patients who received primary prophylaxis (PP group) were compared to patients who received secondary or no G-CSF (no-PP group). Overall survival (OS) and progression-free survival (PFS) were evaluated using the standard Cox proportional hazard model. To account for potential biases, we performed sensitivity analyses excluding patients who received secondary prophilaxis and treating G-CSF as a time-dependent covariate in extended Cox proportional hazard models. RESULTS The study population consisted of 123 patients. PP was used by 75 patients (61.0%). G3/4 N occurred more frequently among patients without PP (10.7 vs 41.7%; P < .001). There was no difference in the frequency of FN between groups (5.3 vs 8.3%; P = .710). In multivariate analysis, PP was associated with a trend toward improved OS (HR = .66; 95% confidence interval [95% CI] .41 - 1.07; P = .094). In the multivariate model excluding patients with secondary prophylaxis (HR = .54; 95% CI 0.32 - .91; P = .022) and in the time-dependent model (HR = .47; 95% CI 0.28 - .80; P = .005), PP was associated with statistically superior OS. CONCLUSIONS Despite the reduction in the frequency of G3/4N, the risk of FN among patients treated with FOLFIRINOX without G-CSF is too low to justify its use in a routine basis. However, given the potential of G-CSF to improve survival in this setting, further studies are warranted to assess its role during treatment with FOLFIRINOX for patients with MPA.
Collapse
Affiliation(s)
| | | | - Victor Hugo Fonseca de Jesus
- Medical Oncology Department, AC Camargo Cancer Center, Sao Paulo, Brazil,Victor Hugo Fonseca de Jesus, MD, MSc, Medical Oncology Department, AC Camargo Cancer Center, Rua Prof. Antonio Prudente, 211, Sao Paulo 01509-010, Brazil.
| |
Collapse
|
15
|
Hegewisch-Becker S, Kratz-Albers K, Wierecky J, Gerhardt S, Reschke D, Borchardt J, Reichelt R, Friedrich FW. Current treatment landscape of pancreatic cancer patients in a network of office-based oncologists in Germany. Future Oncol 2022; 18:3827-3838. [PMID: 36533962 DOI: 10.2217/fon-2022-0141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background: Pancreatic cancer is one of the most aggressive cancers, with comparatively poor outcomes despite the use of multiagent conventional chemotherapy regimens. Real-world data from clinical practice are still rare but are the basis for understanding and improving the current standard of care. Materials & methods: In this multi-institutional retrospective analysis of 24 office-based oncology practices in Germany, the authors documented 1786 pancreatic cancer patients who received systemic treatment between April 2017 and June 2021. Results: The authors' analysis showed that results from recent clinical studies are promptly incorporated into practice. Conclusion: It was striking that, during the analyzed period, the use of platinum-based therapy regimens in adjuvant and palliative first-line therapy increased predominantly in younger patients (<70 years).
Collapse
Affiliation(s)
| | | | - Jan Wierecky
- Überörtliche Gemeinschaftspraxis Schwerpunkt Hämatologie, Onkologie und Palliativmedizin, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
16
|
Agostini A, Orlacchio A, Carbone C, Guerriero I. Understanding Tricky Cellular and Molecular Interactions in Pancreatic Tumor Microenvironment: New Food for Thought. Front Immunol 2022; 13:876291. [PMID: 35711414 PMCID: PMC9193393 DOI: 10.3389/fimmu.2022.876291] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/29/2022] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents 90% of all pancreatic cancer cases and shows a high mortality rate among all solid tumors. PDAC is often associated with poor prognosis, due to the late diagnosis that leads to metastasis development, and limited efficacy of available treatments. The tumor microenvironment (TME) represents a reliable source of novel targets for therapy, and even if many of the biological interactions among stromal, immune, and cancer cells that populate the TME have been studied, much more needs to be clarified. The great limitation in the efficacy of current standard chemoterapy is due to both the dense fibrotic inaccessible TME barrier surrounding cancer cells and the immunological evolution from a tumor-suppressor to an immunosuppressive environment. Nevertheless, combinatorial therapies may prove more effective at overcoming resistance mechanisms and achieving tumor cell killing. To achieve this result, a deeper understanding of the pathological mechanisms driving tumor progression and immune escape is required in order to design rationale-based therapeutic strategies. This review aims to summarize the present knowledge about cellular interactions in the TME, with much attention on immunosuppressive functioning and a specific focus on extracellular matrix (ECM) contribution.
Collapse
Affiliation(s)
- Antonio Agostini
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Arturo Orlacchio
- NYU Grossman School of Medicine, NYU Langone Health, New York, NY, United States
| | - Carmine Carbone
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ilaria Guerriero
- Biogem, Biology and Molecular Genetics Institute, Ariano Irpino, Italy
| |
Collapse
|
17
|
Lee LD, Pozios I, Liu V, Nachbichler SB, Böhmer D, Kamphues C, Beyer K, Bruns CJ, Kreis ME, Seeliger H. Thymidine phosphorylase induction by ionizing radiation antagonizes 5-fluorouracil resistance in human ductal pancreatic adenocarcinoma. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2022; 61:255-262. [PMID: 35084511 PMCID: PMC9021112 DOI: 10.1007/s00411-022-00962-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 01/09/2022] [Indexed: 06/14/2023]
Abstract
Chemoresistance in pancreatic ductal adenocarcinoma (PDAC) frequently contributes to failure of systemic therapy. While the radiosensitizing properties of 5-fluorouracil (FU) are well known, it is unknown whether ionizing radiation (IR) sensitizes towards FU cytotoxicity. Here, we hypothesize that upregulation of thymidine phosphorylase (TP) by IR reverses FU chemoresistance in PDAC cells. The FU resistant variant of the human PDAC cell line AsPC-1 (FU-R) was used to determine the sensitizing effects of IR. Proliferation rates of FU sensitive parental (FU-S) and FU-R cells were determined by WST-1 assays after low (0.05 Gy) and intermediate dose (2.0 Gy) IR followed by FU treatment. TP protein expression in PDAC cells before and after IR was assessed by Western blot. To analyze the specificity of the FU sensitizing effect, TP was ablated by siRNA. FU-R cells showed a 2.7-fold increase of the half maximal inhibitory concentration, compared to FU-S parental cells. Further, FU-R cells showed a concomitant IR resistance towards both doses applied. When challenging both cell lines with FU after IR, FU-R cells had lower proliferation rates than FU-S cells, suggesting a reversal of chemoresistance by IR. This FU sensitizing effect was abolished when TP was blocked by anti-TP siRNA before IR. An increase of TP protein expression was seen after both IR doses. Our results suggest a TP dependent reversal of FU-chemoresistance in PDAC cells that is triggered by IR. Thus, induction of TP expression by low dose IR may be a therapeutic approach to potentially overcome FU chemoresistance in PDAC.
Collapse
Affiliation(s)
- Lucas D Lee
- Department of General and Visceral Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200, Berlin, Germany
| | - Ioannis Pozios
- Department of General and Visceral Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200, Berlin, Germany
| | - Verena Liu
- Department of General and Visceral Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200, Berlin, Germany
| | - Silke B Nachbichler
- Department of Radiotherapy and Radiation Oncology, Klinikum der Universität München, 81377, Munich, Germany
| | - Dirk Böhmer
- Department of Radiation Oncology and Radiotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200, Berlin, Germany
| | - Carsten Kamphues
- Department of General and Visceral Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200, Berlin, Germany
| | - Katharina Beyer
- Department of General and Visceral Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200, Berlin, Germany
| | - Christiane J Bruns
- Department of Surgery, University Hospital of Cologne, 50937, Cologne, Germany
| | - Martin E Kreis
- Department of General and Visceral Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200, Berlin, Germany
| | - Hendrik Seeliger
- Department of General and Visceral Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200, Berlin, Germany.
- IU Health University, 10243, Berlin, Germany.
| |
Collapse
|
18
|
Modified FOLFIRINOX as a Second-Line Treatment for Patients with Gemcitabine-Failed Advanced Biliary Tract Cancer: A Prospective Multicenter Phase II Study. Cancers (Basel) 2022; 14:cancers14081950. [PMID: 35454857 PMCID: PMC9029419 DOI: 10.3390/cancers14081950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/10/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Biliary tract cancer is a malignant tumor of the biliary tract and gallbladder. Most patients are diagnosed at an advanced stage, and the basis of treatment is combination chemotherapy. However, the survival outcomes for biliary tract cancer, especially for patients who have failed frontline treatment, are poor. Accordingly, there have been various studies on effective subsequent treatments, and this study is one of those efforts. Through this study, we attempted to demonstrate the efficacy of enhanced chemotherapy in relapsed or refractory biliary tract cancer. Abstract Background: After the publication of the ABC-02 trial, gemcitabine and cisplatin combination therapy (GP) became the standard first-line treatment for advanced biliary tract cancer (BTC). Despite GP therapy, most patients suffer from disease progression. The ABC-06 trial recommended FOLFOX as a second-line treatment, but its efficacy was modest. In this phase II study, we looked at the efficacy and safety of a second-line modified dose of FOLFIRINOX (mFOLFIRINOX) for patients who had failed first-line gemcitabine-based treatment. Methods: From January 2020 to January 2021, 34 patients with advanced BTC who failed first-line gemcitabine-based chemotherapy were enrolled. We evaluated the clinical efficacy and safety outcomes of mFOLFIRINOX. Results: With a median follow-up duration of 13.4 months, the median progression-free survival and overall survival was 2.8 months (95% confidence interval (CI): 1.6–4.0 months) and 6.2 months (95% CI: 5.0–7.4 months), respectively. The objective response rate was 14.7% with no complete response. The disease control rate was 61.7%, with a disease control duration of 4.2 months. Due to the rapid progression of the disease, approximately half of all patients received less than three cycles of treatment. The most common type of adverse event (AEs) was hematopoietic AEs. The incidence of non-hematopoietic AEs was relatively low. Conclusions: The efficacy of mFOLFIRINOX as a second-line treatment in advanced BTC patients after the failure of gemcitabine-based first-line treatment was replicated, albeit with slightly shorter survival results compared to previous studies. Long-term administration of mFOLFIRINOX with toxicity management might offer a survival benefit.
Collapse
|
19
|
Sugimoto M, Takagi T, Suzuki R, Konno N, Asama H, Sato Y, Irie H, Okubo Y, Nakamura J, Takasumi M, Hashimoto M, Kato T, Kobashi R, Hikichi T, Ohira H. Drug treatment for chemotherapy-induced peripheral neuropathy in patients with pancreatic cancer. Fukushima J Med Sci 2022; 68:1-10. [PMID: 35197393 PMCID: PMC9071352 DOI: 10.5387/fms.2021-32] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/11/2022] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer (PC) is a lethal disease where most tumors are too advanced at diagnosis for resection, leaving chemotherapy as the mainstay of treatment. Although the prognosis of unresectable PC is poor, it has been dramatically improved by new chemotherapy treatments, such as the combination of 5-fluorouracil, oxaliplatin, irinotecan, and leucovorin (FOLFIRINOX) or gemcitabine plus nab-paclitaxel. However, as oxaliplatin and paclitaxel are common neurotoxic drugs, chemotherapy-induced peripheral neuropathy (CIPN) is a common and severe adverse effect of both treatments. As there are no agents recommended in the ASCO guidelines, we review the methods used to treat CIPN caused by PC treatment. The efficacy of duloxetine was observed in a large randomized controlled trial (RCT). In addition, pregabalin was more effective than duloxetine for CIPN in two RCTs. Although duloxetine and pregabalin can be effective for CIPN, they have several side effects. Therefore, the choice between the two drugs should be determined according to effect and tolerability. Mirogabalin is also used in patients with PC and there is hope it will yield positive outcomes when treating CIPN in the future.
Collapse
Affiliation(s)
- Mitsuru Sugimoto
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
| | - Tadayuki Takagi
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
| | - Rei Suzuki
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
| | - Naoki Konno
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
| | - Hiroyuki Asama
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
| | - Yuki Sato
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
| | - Hiroki Irie
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
| | - Yoshinori Okubo
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
- Department of Endoscopy, Fukushima Medical University Hospital
| | - Jun Nakamura
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
- Department of Endoscopy, Fukushima Medical University Hospital
| | - Mika Takasumi
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
| | - Minami Hashimoto
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
- Department of Endoscopy, Fukushima Medical University Hospital
| | - Tsunetaka Kato
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
- Department of Endoscopy, Fukushima Medical University Hospital
| | - Ryoichiro Kobashi
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
| | - Takuto Hikichi
- Department of Endoscopy, Fukushima Medical University Hospital
| | - Hiromasa Ohira
- Department of Gastroenterology, School of Medicine, Fukushima Medical University
| |
Collapse
|
20
|
Shankara Narayanan JSN, Frizzi K, Erdem S, Ray P, Jaroch D, Cox B, Katz S, Vicente D, White R. Oxaliplatin-induced peripheral neuropathy can be minimized by pressurized regional intravascular delivery in an orthotopic murine pancreatic cancer model. Discov Oncol 2022; 13:21. [PMID: 35384564 PMCID: PMC8986945 DOI: 10.1007/s12672-022-00483-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/23/2022] [Indexed: 12/03/2022] Open
Abstract
PURPOSE There is a great need to reduce the toxicity of chemotherapy used in the management of pancreatic ductal adenocarcinoma (PDAC). Here we explore if regional pressurized delivery of oxaliplatin can minimize peripheral neuropathy in mice. METHODS We used an orthotopic PDAC mouse model and delivered a single dose of oxaliplatin through the portal vein using a pressure-enabled system (pancreatic retrograde venous infusion, PRVI). We analyzed the effects of PRVI on tumor burden and peripheral neuropathy using histopathological and functional assays. RESULTS Tumor weights in mice treated with 2 mg/kg oxaliplatin using PRVI were significantly lower than in mice treated with the same dose systemically. This resulted in reduced peripheral neuropathy signatures in PRVI mice compared to the 20 mg/kg systemic dose required to achieve similar tumor control. CONCLUSION Regional delivery of highly cytotoxic agents using PRVI can reduce the therapeutic dose of these drugs, thereby lowering toxic side effects.
Collapse
Affiliation(s)
| | - Katie Frizzi
- Department of Pathology, University of California, San Diego, CA, USA
| | - Suna Erdem
- Moores Cancer Center, University of California, 3855 Health Sciences Dr, Rm 2336, La Jolla, San Diego, CA, 92037, USA
| | - Partha Ray
- Moores Cancer Center, University of California, 3855 Health Sciences Dr, Rm 2336, La Jolla, San Diego, CA, 92037, USA
| | - David Jaroch
- TriSalus™ Life Sciences, Inc, Westminster, CO, USA
| | - Bryan Cox
- TriSalus™ Life Sciences, Inc, Westminster, CO, USA
| | - Steven Katz
- TriSalus™ Life Sciences, Inc, Westminster, CO, USA
- Immuno-Oncology Institute, Roger Williams Medical Center, Providence, RI, USA
| | - Diego Vicente
- Moores Cancer Center, University of California, 3855 Health Sciences Dr, Rm 2336, La Jolla, San Diego, CA, 92037, USA
- Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Rebekah White
- Moores Cancer Center, University of California, 3855 Health Sciences Dr, Rm 2336, La Jolla, San Diego, CA, 92037, USA.
| |
Collapse
|
21
|
Ettrich TJ, Sturm N, Güthle M, Hüttner FJ, Perkhofer L. Pancreatic Cancer: Current Multimodality Treatment Options and the Future Impact of Molecular Biological Profiling. Visc Med 2022; 38:20-29. [PMID: 35295894 PMCID: PMC8874237 DOI: 10.1159/000521631] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/19/2021] [Indexed: 02/03/2023] Open
Abstract
Background Pancreatic cancer (PDAC) - even if deemed resectable - has still a dismal prognosis and is the seventh leading cause of global cancer-related death with rising incidence worldwide. Summary Surgical resection at best in combination with adjuvant systemic chemotherapy is the only potentially curative treatment. Surgical treatment has substantially improved over the last years with significantly reduced perioperative morbidity and mortality. Even when deemed radiologically resectable, the majority of PDAC is likely to have micrometastases, leaving most PDAC patients with an advanced stage. Recent 5-year overall survival was up to 46% in patients eligible for surgery with intensified adjuvant chemotherapy. Eligible for curative surgery are about one-third of the patients, and only 20% of these patients have the option for cure with surgery and adjuvant chemotherapy. Standards of care in treating PDAC patients include various mostly combinational chemotherapy approaches in the advanced and adjuvant setting. Moreover, first targeted therapies for individualizing treatment, e.g., specific subgroups like BRCA1/2 germline mutated patients, were established lately. Neoadjuvant concepts are currently part of research. This review focuses on current and future multimodal treatment options of PDAC and the impact of molecular profiling for individualizing treatment. Key Messages State of the art in pancreatic cancer therapy is multimodal and includes novel strategies to allow molecular defined subgroup-specific treatment.
Collapse
Affiliation(s)
- Thomas J. Ettrich
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany,*Thomas J. Ettrich,
| | - Niklas Sturm
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Melanie Güthle
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Felix J. Hüttner
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Lukas Perkhofer
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
22
|
Queck A, Elango S, Koch C, Walter D, Schmidt J, Trebicka J, Trojan J, Pession U, Finkelmeier F, Waidmann O. Sequence Therapy with FOLFIRINOX and Gemcitabine/Nab-Paclitaxel for Patients with Advanced Pancreatic Cancer - a Monocenter Retrospective Cohort Study. Oncol Res Treat 2021; 45:79-87. [PMID: 34875671 DOI: 10.1159/000521258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/30/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND & AIMS While irresectable pancreatic cancer has still a dismal overall prognosis, evidence about the optimal chemotherapy sequence is scarce. After treatment with FOLFIRINOX in first-line, Gemcitabine-monotherapy was established for years. As a potential treatment alternative after failure of FOLFIRINOX therapy, combination of Gemcitabine and Nab-Paclitaxel is used. However, this combination has formally not yet been approved for second-line treatment and investigation of efficiency and treatment tolerance is the aim of this trial. METHODS Therefore, we investigated 225 patients with histologically confirmed local advanced or metastatic pancreatic cancer in this retrospective mono-centre study (November 2010 - July 2019). Of this, 44 patients received FOLFIRINOX therapy and outcome was further analysed. The primary end point of this cohort was overall survival, secondary end points included progression free survival, response rate, and safety. RESULTS In most of the patients FOLFIRINOX as first-line treatment of irresectable pancreatic cancer resulted in temporary cancer control (partial response (PR): 50% and stable disease (SD): 18%), whereas tumor progression was observed in 23% of the patients. The median progression-free survival (PFS) time for FOLFIRINOX treatment was 7.3 months and median overall survival 10.3 months. Seven (16%) patients received additional local radio chemotherapy of the pancreatic tumor. During first-line therapy 8 (18%) patients had laparotomy for proof of resectability. Hereby, in three patients R0-, in three patients R1 resection, and irresectability in another 2 patients were achieved. Twenty-five of the 44 patients (57%) received second line therapy, namely 24 patients Gemcitabine/ Nab-Paclitaxel and 1 patient Gemcitabine and Erlotinib. Hereby, Gemcitabine/ Nab-Paclitaxel led again to temporary tumor control in 46% of the patients (PR: 21%, SD: 25%), while in 29% of the patient's disease progression was observed. Corresponding median PFS for Gemcitabine and Nab-Paclitaxel treatment was 3.5 months. Patients who received second-line treatment with Nab-Paclitaxel and Gemcitabine had a more favorable prognosis (median OS: 17.4 versus 9.2 months; HR 0.32 [0.14 - 0.70], p<0.001) than patients who were not eligible for second-line treatment. Moreover, in multivariate analyses association with patients' survival and tumor response to chemotherapy in both therapeutic lines and µGT below 100 IU/L in first-line FOLFIRINOX chemotherapy were observed. CONCLUSION These real-world data suggest that Gemcitabine / Nab-Paclitaxel may be feasible after FOLFIRINOX therapy in patients with irresectable pancreatic cancer. However, prospective randomized data about the superiority to Gemcitabine monotherapy are needed.
Collapse
Affiliation(s)
- Alexander Queck
- Department of Gastroenterology, Hepatology and Endocrinology, University Hospital Frankfurt, Frankfurt, Germany,
| | - Sharra Elango
- Department of Gastroenterology, Hepatology and Endocrinology, University Hospital Frankfurt, Frankfurt, Germany
| | - Christine Koch
- Department of Gastroenterology, Hepatology and Endocrinology, University Hospital Frankfurt, Frankfurt, Germany
- University Cancer Center Frankfurt, Frankfurt, Germany
| | - Dirk Walter
- Department of Gastroenterology, Hepatology and Endocrinology, University Hospital Frankfurt, Frankfurt, Germany
- University Cancer Center Frankfurt, Frankfurt, Germany
| | - Jennifer Schmidt
- Department of Gastroenterology, Hepatology and Endocrinology, University Hospital Frankfurt, Frankfurt, Germany
- University Cancer Center Frankfurt, Frankfurt, Germany
| | - Jonel Trebicka
- Department of Gastroenterology, Hepatology and Endocrinology, University Hospital Frankfurt, Frankfurt, Germany
| | - Jörg Trojan
- Department of Gastroenterology, Hepatology and Endocrinology, University Hospital Frankfurt, Frankfurt, Germany
- University Cancer Center Frankfurt, Frankfurt, Germany
| | - Ursula Pession
- University Cancer Center Frankfurt, Frankfurt, Germany
- Department of General, Visceral and Transplant Surgery, University Hospital Frankfurt, Frankfurt, Germany
| | - Fabian Finkelmeier
- Department of Gastroenterology, Hepatology and Endocrinology, University Hospital Frankfurt, Frankfurt, Germany
- University Cancer Center Frankfurt, Frankfurt, Germany
| | - Oliver Waidmann
- Department of Gastroenterology, Hepatology and Endocrinology, University Hospital Frankfurt, Frankfurt, Germany
- University Cancer Center Frankfurt, Frankfurt, Germany
| |
Collapse
|
23
|
Abstract
Pancreatic cancer is mainly diagnosed at an advanced, often metastatic stage and still has a poor prognosis. Over the last decades, chemotherapy of metastatic pancreatic cancer (mPDAC) has proven to be superior to a mere supportive treatment with respect to both survival and quality of life. Recently, even sequential treatment of mPDAC could be established. Options for first-line treatment are combination chemotherapy regimens such as FOLFIRINOX and gemcitabine plus nab-paclitaxel when the performance status of the patient is good. For patients with poorer performance status, gemcitabine single-agent treatment is a valid option. Recently, the PARP inhibitor olaparib has been demonstrated to improve progression-free survival when used as a maintenance treatment in the subgroup of patients with mPDAC and a BRCA1/-2 germ line mutation having received at least 16 weeks of platinum-based chemotherapy. This group of patients also benefits from platinum-based chemotherapy combinations. Therefore, the BRCA1/-2 stats should be examined early in patients with mPDAC even when the occurrence of these mutations is only about 5% in the general Caucasian population. After the failure of first-line treatment, patients should be offered a second-line treatment if their ECOG permits further treatment. Here, the combination of 5-FU/FA plus nanoliposomal irinotecan has shown to be superior to 5-FU/FA alone with respect to overall survival. Immune checkpoint inhibitors like PD1/PD-L1 mAbs are particularly efficacious in tumors with high microsatellite instability (MSI-h). Limited data in mPDACs shows that only a part of the already small subgroup of MSI-H mPDACs (frequency about 1%) appears to benefit substantially from a checkpoint inhibitor treatment. The identification of further subgroups, e.g., tumors with DNA damage repair deficiency, gene fusions, as well as novel approaches such as tumor-organoid-informed treatment decisions, may further improve therapeutic efficacy.
Collapse
|
24
|
Harada S, Iida T, Asai S, Nakamura K, Ishihara M, Iwasaki J, Itami A, Kyogoku T. Successful En Bloc Resection of Locally Advanced Pancreatic Tail Cancer with Colonic Perforation Following Neoadjuvant Chemotherapy: A Case Report. AMERICAN JOURNAL OF CASE REPORTS 2021; 22:e933226. [PMID: 34650027 PMCID: PMC8525901 DOI: 10.12659/ajcr.933226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Distal pancreatic cancers may be unresectable at the time of diagnosis because these cancers are asymptomatic and readily infiltrate neighboring organs. Radical resection of a pancreatic tail cancer with colonic perforation is rare. We describe successful resection of a locally advanced pancreatic tail cancer with colonic perforation using a multidisciplinary approach. CASE REPORT A 66-year-old man presented to our hospital with a chief concern of high fever. Abdominal computed tomography revealed a pancreatic tail tumor infiltrating the neighboring organs and causing colonic obstruction with perforation, which resulted in an intra-abdominal abscess. Colonoscopy revealed obstruction of the descending colon by extramural invasion. Laboratory tests showed high tumor marker concentrations (carcinoembryonic antigen, 11.6 ng/dL; pancreatic cancer-associated antigen-2, >1600 U/mL). We clinically diagnosed locally advanced pancreatic tail cancer with an intra-abdominal abscess caused by colonic perforation. First, we performed transverse colostomy and percutaneous drainage. We then started neoadjuvant chemotherapy with FOLFIRINOX for tumor shrinkage and prevention of distant metastases. The therapeutic effect was a partial response, and no distant metastases was found. We therefore performed radical surgery comprising distal pancreatectomy with partial resection of neighboring organs. Although pathological examination revealed a pancreatic tail tubular adenocarcinoma with direct invasion of the neighboring organs, R0 resection was achieved. The patient was discharged with no perioperative complications. Tegafur/gimeracil/oteracil potassium were administered as adjuvant chemotherapy. The patient remained recurrence-free for 19 months after surgery. CONCLUSIONS We achieved successful en bloc resection of a locally advanced distal pancreatic cancer with colonic perforation by using a multidisciplinary approach.
Collapse
Affiliation(s)
- Shigeyuki Harada
- Department of Surgery, Kobe City Nishi-Kobe Medical Center, Kobe, Hyogo, Japan
| | - Taku Iida
- Department of Surgery, Kobe City Nishi-Kobe Medical Center, Kobe, Hyogo, Japan
| | - Satsuki Asai
- Department of Pathology, Kobe City Nishi-Kobe Medical Center, Kobe, Hyogo, Japan
| | - Kojiro Nakamura
- Department of Surgery, Kobe City Nishi-Kobe Medical Center, Kobe, Hyogo, Japan
| | - Misa Ishihara
- Department of Pathology, Kobe City Nishi-Kobe Medical Center, Kobe, Hyogo, Japan
| | - Junji Iwasaki
- Department of Surgery, Kobe City Nishi-Kobe Medical Center, Kobe, Hyogo, Japan
| | - Atsushi Itami
- Department of Surgery, Kobe City Nishi-Kobe Medical Center, Kobe, Hyogo, Japan
| | - Takahisa Kyogoku
- Department of Surgery, Kobe City Nishi-Kobe Medical Center, Kobe, Hyogo, Japan
| |
Collapse
|
25
|
Use of FOLFIRINOX or Nab-Paclitaxel Plus Gemcitabine for the Treatment of Locally Advanced Pancreatic Adenocarcinoma: A Single Institution Observational Study. Cancers (Basel) 2021; 13:cancers13194939. [PMID: 34638422 PMCID: PMC8508515 DOI: 10.3390/cancers13194939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary We performed a retrospective analysis to evaluate the effect of treatment with FOLFIRINOX (FFN) or Nab-paclitaxel plus gemcitabine (GemNab) in patients with locally advanced (LA) pancreatic adenocarcinoma (PDAC). Forty-eight percent of patients treated with FFN became eligible for radical resection, and twenty-two percent of patients receiving GemNab underwent surgery after neoadjuvant treatment. FFN treatment was associated with a better overall survival, compared with GemNab (mOS 85.1 vs. 54.3 weeks, FFN and GemNab, respectively; HR = 0.54; p = 0.0109). We found different toxicity profiles between the two chemotherapy regimens. Future randomized clinical trials are mandatory to clarify the best treatment in patients with LA PDAC. Abstract Patients with locally advanced (LA) pancreatic ductal adenocarcinoma (PDAC) do not present distant metastases but are not eligible for surgery upfront. Chemotherapy regimens, such as FOLFIRINOX (FFN) or nab-paclitaxel plus gemcitabine (GemNab) in combination with loco-regional treatments are generally used in this setting. However, the best treatment choice is unknown. We retrospectively analyzed the information of 225 patients with stage II–III PDAC treated at our institution between October 2011 and December 2020. A total of 94 patients with LA PDAC who are non-eligible for surgery upfront received neoadjuvant FFN or GemNab. Of the 67 patients receiving FFN, 28 (41.8%) underwent surgery after neoadjuvant therapy. Of the 27 patients treated with GemNab, 6 (22.2%) became eligible for resection. The median overall survival (OS) was 85.1 weeks and 54.3 weeks in the FFN and GemNab groups, respectively (HR = 0.54, p = 0.0109). The median OS was 189.7 weeks and 76.4 weeks in the resected and unresected cohorts, respectively (HR = 0.25, p < 0.0001). Neutropenia (37.3%), anemia (6.0%), and diarrhea (6.0%) in the FFN group and neutropenia (22.2%) and thrombocytopenia (18.5%) in the GemNab groups were the most frequent grade 3–4 side effects. Higher rates of thrombocytosis (p < 0.0001) and peripheral edema (p < 0.0001) were observed in the GemNab group. Our results suggest that the use of FFN is associated with more favorable clinical outcomes than GemNab for patients with LA PDAC. Future randomized and controlled clinical trials are needed to further elucidate the role of these regimens and loco-regional treatments in this setting.
Collapse
|
26
|
Mollinedo F, Gajate C. Direct Endoplasmic Reticulum Targeting by the Selective Alkylphospholipid Analog and Antitumor Ether Lipid Edelfosine as a Therapeutic Approach in Pancreatic Cancer. Cancers (Basel) 2021; 13:4173. [PMID: 34439330 PMCID: PMC8394177 DOI: 10.3390/cancers13164173] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/11/2021] [Accepted: 08/15/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common malignancy of the pancreas, shows a dismal and grim overall prognosis and survival rate, which have remained virtually unchanged for over half a century. PDAC is the most lethal of all cancers, with the highest mortality-to-incidence ratio. PDAC responds poorly to current therapies and remains an incurable malignancy. Therefore, novel therapeutic targets and drugs are urgently needed for pancreatic cancer treatment. Selective induction of apoptosis in cancer cells is an appealing approach in cancer therapy. Apoptotic cell death is highly regulated by different signaling routes that involve a variety of subcellular organelles. Endoplasmic reticulum (ER) stress acts as a double-edged sword at the interface of cell survival and death. Pancreatic cells exhibit high hormone and enzyme secretory functions, and thereby show a highly developed ER. Thus, pancreatic cancer cells display a prominent ER. Solid tumors have to cope with adverse situations in which hypoxia, lack of certain nutrients, and the action of certain antitumor agents lead to a complex interplay and crosstalk between ER stress and autophagy-the latter acting as an adaptive survival response. ER stress also mediates cell death induced by a number of anticancer drugs and experimental conditions, highlighting the pivotal role of ER stress in modulating cell fate. The alkylphospholipid analog prototype edelfosine is selectively taken up by tumor cells, accumulates in the ER of a number of human solid tumor cells-including pancreatic cancer cells-and promotes apoptosis through a persistent ER-stress-mediated mechanism both in vitro and in vivo. Here, we discuss and propose that direct ER targeting may be a promising approach in the therapy of pancreatic cancer, opening up a new avenue for the treatment of this currently incurable and deadly cancer. Furthermore, because autophagy acts as a cytoprotective response to ER stress, potentiation of the triggering of a persistent ER response by combination therapy, together with the use of autophagy blockers, could improve the current gloomy expectations for finding a cure for this type of cancer.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, C/Ramiro de Maeztu 9, E-28040 Madrid, Spain;
| | | |
Collapse
|
27
|
Chen J, Hua Q, Wang H, Zhang D, Zhao L, Yu D, Pi G, Zhang T, Lin Z. Meta-analysis and indirect treatment comparison of modified FOLFIRINOX and gemcitabine plus nab-paclitaxel as first-line chemotherapy in advanced pancreatic cancer. BMC Cancer 2021; 21:853. [PMID: 34301232 PMCID: PMC8306351 DOI: 10.1186/s12885-021-08605-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/16/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Modified FOLFIRINOX and gemcitabine plus nab-paclitaxel (GEM-NAB) have been recommended as first-line therapies for advanced pancreatic cancer (PC). Due to the lack of evidence to directly compare them, we conducted this network meta-analysis to indirectly compare the effectiveness and toxicity of modified FOLFIRINOX and GEM-NAB. METHODS The eligible retrospective studies on treatments related to modified FOLFIRINOX and GEM-NAB up to 4 April 2020 were searched and assessed. We used the frequentist model to analyze the survival and toxicity data between different treatments. Pooled analysis for overall survival (OS), progression-free survival (PFS), objective response rate (ORR) and events of toxicity were analyzed in this study. RESULTS Twenty-two studies were involved in this network meta-analysis. The comparisons on OS and PFS showed that modified FOLFIRINOX and GEM-NAB had similar treatment efficacy (OS: 1.13; 95% CI: 0.78-1.63; PFS: HR: 1.19; 95% CI: 0.85-1.67). GEM-NAB was more effective than modified FOLFIRINOX based on the result of ORR (RR: 1.43; 95% CI: 1.04-1.96). Moreover, our analysis showed a similar toxicity profile between modified FOLFIRINOX and GEM-NAB. CONCLUSIONS The current evidence showed that modified FOLFIRINOX and GEM-NAB were similar in survival and toxicity. Many factors should be considered for in the formulation of optimal treatment, and our meta-analysis could provide some guidance to treatment selection in the first-line setting for advanced PC.
Collapse
Affiliation(s)
- Jiayuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qingling Hua
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Haihong Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dejun Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lei Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dandan Yu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guoliang Pi
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhenyu Lin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
28
|
Nesbitt H, Logan K, Thomas K, Callan B, Gao J, McKaig T, Taylor M, Love M, Stride E, McHale AP, Callan JF. Sonodynamic therapy complements PD-L1 immune checkpoint inhibition in a murine model of pancreatic cancer. Cancer Lett 2021; 517:88-95. [PMID: 34119606 DOI: 10.1016/j.canlet.2021.06.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 02/07/2023]
Abstract
The emergence of immune checkpoint inhibitors (ICI's) in the past decade has proven transformative in the area of immuno-oncology. The PD-1/PD-L1 axis has been particularly well studied and monoclonal antibodies developed to block either the receptor (anti PD-1) or its associated ligand (anti PD-L1) can generate potent anti-tumour immunity in certain tumour models. However, many "immune cold" tumours remain unresponsive to ICI's and strategies to stimulate the adaptive immune system and make these tumours more susceptible to ICI treatment are currently under investigation. Sonodynamic therapy (SDT) is a targeted anti-cancer treatment that uses ultrasound to activate a sensitiser with the resulting generation of reactive oxygen species (ROS) causing direct cell death by apoptosis and necrosis. SDT has also been shown to stimulate the adaptive immune system in a pre-clinical model of colorectal cancer. In this manuscript, we investigate the ability of microbubble mediated SDT to control tumour growth in a bilateral tumour mouse model of pancreatic cancer by treating the target tumour with SDT and observing the effects at the off-target untreated tumour. The results demonstrated a significant 287% decrease in tumour volume when compared to untreated animals 11 days following the initial treatment with SDT, which reduced further to 369% when SDT was combined with anti-PD-L1 ICI treatment. Analysis of residual tumour tissues remaining after treatment revealed increased levels of infiltrating CD4+ and CD8+ T-lymphocytes (respectively 4.65 and 3.16-fold more) in the off-target tumours of animals where the target tumour was treated with SDT and anti-PD-L1, when compared to untreated tumours. These results suggest that SDT treatment elicits an adaptive immune response that is potentiated by the anti-PD-L1 ICI in this particular model of pancreatic cancer.
Collapse
Affiliation(s)
- Heather Nesbitt
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, UK
| | - Keiran Logan
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, UK
| | - Keith Thomas
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, UK
| | - Bridgeen Callan
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, UK
| | - Jinhui Gao
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, UK
| | - Thomas McKaig
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, UK
| | - Mark Taylor
- Department of HPB Surgery, Mater Hospital, Belfast, Northern Ireland, BT14 6AB, UK
| | - Mark Love
- Imaging Centre, The Royal Victoria Hospital, Grosvenor Road, Belfast, Northern Ireland, BT12 6BA, UK
| | - Eleanor Stride
- Institute of Biomedical Engineering, University of Oxford, Oxford, OX3 7DQ, UK
| | - Anthony P McHale
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, UK
| | - John F Callan
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, UK.
| |
Collapse
|
29
|
Innominato PF, Karaboué A, Focan C, Chollet P, Giacchetti S, Bouchahda M, Ulusakarya A, Torsello A, Adam R, Lévi FA, Garufi C. Efficacy and safety of chronomodulated irinotecan, oxaliplatin, 5-fluorouracil and leucovorin combination as first- or second-line treatment against metastatic colorectal cancer: Results from the International EORTC 05011 Trial. Int J Cancer 2021; 148:2512-2521. [PMID: 33270911 PMCID: PMC8048520 DOI: 10.1002/ijc.33422] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/20/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
The triplet combination of irinotecan, oxaliplatin and fluorouracil is an active frontline regimen in metastatic colorectal cancer, but scarce data exist on its use as salvage treatment. We aimed at assessing its safety and efficacy profiles with its circadian-based administration (chronoIFLO5) as either first- or second-line treatment, within the time-finding EORTC 05011 trial. Five-day chronoIFLO5 was administered every 3 weeks in patients with PS 0, 1 or 2. It consisted of chronomodulated irinotecan (180 mg/sqm), oxaliplatin (80 mg/sqm) and fluorouracil-leucovorin (2800 and 1200 mg/sqm, respectively). For our study, toxicity and antitumour activity were evaluated separately in first- and second-line settings. Primary endpoints included Grade 3-4 toxicity rates, best objective response rate (ORR), progression-free survival (PFS) and overall survival (OS). One-hundred forty-nine and 44 patients were treated in first-line and second-line settings, respectively, with a total of 1138 cycles with median relative dose intensities of about 90%. Demographics were comparable in the two groups. Thirty-six (24.7%) and 10 (22.2%) patients experienced at least one episode of severe toxicity in first line and second line, respectively. Frontline chronoIFLO5 yielded an ORR of 62.3% [95% CI: 54.2-70.4] and resulted in median PFS and OS of 8.7 months [7.5-9.9] and 19.9 months [15.4-24.5]. Corresponding figures in second line were 37.5% [22.5-52.5], 6.7 months [4.8-8.9] and 16.3 months [11.8-20.8]. International and prospective evaluation revealed the favourable safety and efficacy profiles of chronoIFLO5, both as frontline and as salvage treatment against metastatic colorectal cancer. In particular, encouraging activity in second line was observed, with limited haematological toxicity.
Collapse
Affiliation(s)
- Pasquale F. Innominato
- North Wales Cancer Centre, Ysbyty Gwynedd, Betsi Cadwaladr University Health BoardBangorUK
- Cancer Chronotherapy Team, Cancer Research Centre, Division of Biomedical SciencesWarwick Medical SchoolCoventryUK
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
| | - Abdoulaye Karaboué
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Medical Oncology UnitGHI Le Raincy‐MontfermeilMontfermeilFrance
| | - Christian Focan
- Department of OncologyCHC‐MontLegia, Groupe Santé CHC‐LiègeLiègeBelgium
| | - Philippe Chollet
- Clinical and Translational Research DivisionJean Perrin Comprehensive Cancer CentreClermont‐FerrandFrance
| | - Sylvie Giacchetti
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Department of OncologySaint Louis Hospital, Public Hospitals of Paris (AP‐HP)ParisFrance
| | - Mohamed Bouchahda
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Medical Oncology UnitClinique du MousseauEvryFrance
- Medical Oncology UnitClinique Saint Jean L'ErmitageMelunFrance
- Chronotherapy Unit, Department of Medical OncologyPaul Brousse Hospital, Public Hospitals of Paris (AP‐HP)VillejuifFrance
| | - Ayhan Ulusakarya
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Chronotherapy Unit, Department of Medical OncologyPaul Brousse Hospital, Public Hospitals of Paris (AP‐HP)VillejuifFrance
| | - Angela Torsello
- Division of Medical OncologySan Giovanni‐ Addolorata HospitalRomeItaly
| | - René Adam
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Hepatobiliary CentrePaul Brousse Hospital, Public Hospitals of Paris (AP‐HP)VillejuifFrance
| | - Francis A. Lévi
- Cancer Chronotherapy Team, Cancer Research Centre, Division of Biomedical SciencesWarwick Medical SchoolCoventryUK
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Hepatobiliary CentrePaul Brousse Hospital, Public Hospitals of Paris (AP‐HP)VillejuifFrance
| | - Carlo Garufi
- Division of Medical OncologySan Camillo Forlanini HospitalRomeItaly
| |
Collapse
|
30
|
Weiler S, Nairz M. TAM-ing the CIA-Tumor-Associated Macrophages and Their Potential Role in Unintended Side Effects of Therapeutics for Cancer-Induced Anemia. Front Oncol 2021; 11:627223. [PMID: 33842333 PMCID: PMC8027083 DOI: 10.3389/fonc.2021.627223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 03/01/2021] [Indexed: 12/21/2022] Open
Abstract
Cancer-induced anemia (CIA) is a common consequence of neoplasia and has a multifactorial pathophysiology. The immune response and tumor treatment, both intended to primarily target malignant cells, also affect erythropoiesis in the bone marrow. In parallel, immune activation inevitably induces the iron-regulatory hormone hepcidin to direct iron fluxes away from erythroid progenitors and into compartments of the mononuclear phagocyte system. Moreover, many inflammatory mediators inhibit the synthesis of erythropoietin, which is essential for stimulation and differentiation of erythroid progenitor cells to mature cells ready for release into the blood stream. These pathophysiological hallmarks of CIA imply that the bone marrow is not only deprived of iron as nutrient but also of erythropoietin as central growth factor for erythropoiesis. Tumor-associated macrophages (TAM) are present in the tumor microenvironment and display altered immune and iron phenotypes. On the one hand, their functions are altered by adjacent tumor cells so that they promote rather than inhibit the growth of malignant cells. As consequences, TAM may deliver iron to tumor cells and produce reduced amounts of cytotoxic mediators. Furthermore, their ability to stimulate adaptive anti-tumor immune responses is severely compromised. On the other hand, TAM are potential off-targets of therapeutic interventions against CIA. Red blood cell transfusions, intravenous iron preparations, erythropoiesis-stimulating agents and novel treatment options for CIA may interfere with TAM function and thus exhibit secondary effects on the underlying malignancy. In this Hypothesis and Theory, we summarize the pathophysiological hallmarks, clinical implications and treatment strategies for CIA. Focusing on TAM, we speculate on the potential intended and unintended effects that therapeutic options for CIA may have on the innate immune response and, consequently, on the course of the underlying malignancy.
Collapse
Affiliation(s)
- Stefan Weiler
- National Poisons Information Centre, Tox Info Suisse, Associated Institute of the University of Zurich, Zurich, Switzerland.,Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Eidgenossische Technische Hochschule Zurich, Zurich, Switzerland
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
31
|
Laser Treatment of Pancreatic Cancer with Immunostimulating Interstitial Laser Thermotherapy Protocol: Safety and Feasibility Results From Two Phase 2a Studies. J Surg Res 2020; 259:1-7. [PMID: 33278792 DOI: 10.1016/j.jss.2020.10.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/03/2020] [Accepted: 10/31/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE Ablative techniques have emerged as new potential therapeutic options for patients with locally advanced pancreatic cancer (LAPC). We explored the safety and feasibility of using TRANBERG|Thermal Therapy System (Clinical Laserthermia Systems AB, Lund, Sweden) in feedback mode for immunostimulating Interstitial Laser Thermotherapy (imILT) protocol, the newest ablative technique introduced for the treatment of LAPC. METHODS The safety and feasibility results after the use of imILT protocol treatment in 15 patients of a prospective series of postsystemic therapy LAPC in two high-volume European institutions, the General and Pancreatic Unit of the Pancreas Institute, of the University of Verona, Italy, and the Department of Surgical Oncology of the Institut Paoli-Calmettes of Marseille, France, were assessed. RESULTS The mean age was 66 ± 5 years, with a mean tumor size of 34.6 (±8) mm. The median number of cycles of pre-imILT chemotherapy was 6 (6-12). The procedure was performed in 13 of 15 (86.6%) cases; indeed, in two cases, the procedure was not performed; in one, the procedure was considered technically demanding; in the other, liver metastases were found intraoperatively. In all treated cases, the procedure was completed. Three late pancreatic fistulas developed over four overall adverse events (26.6%) and were attributed to imILT. Mortality was nil. A learning curve is necessary to interpret and manage the laser parameters. CONCLUSIONS Safety, feasibility, and device handling outcomes of using TRANBERG|Thermal Therapy System with temperature probes in feedback mode and imILT protocol on LAPC were not satisfactory. The metastatic setting may be appropriate to evaluate the hypothetic abscopal effect.#NCT02702986 and #NCT02973217.
Collapse
|
32
|
Observational Study of Clinical Practice in Patients with Pancreatic Adenocarcinoma in Greece. JOURNAL OF ONCOLOGY 2020; 2020:5304516. [PMID: 33014051 PMCID: PMC7520678 DOI: 10.1155/2020/5304516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 08/14/2020] [Accepted: 09/07/2020] [Indexed: 11/21/2022]
Abstract
Background During the last decade, significant improvement was made in systemic therapy of pancreatic adenocarcinoma (PAC). The impact of this progress in everyday clinical practice has not been fully described yet. The aim of the study was to investigate the pattern followed by Greek Medical Oncologists regarding the treatment of patients with PAC. Methods This observational, noninterventional multicenter study recorded clinical data from the files of 200 active patients (alive and under treatment or follow-up) for a two-year period (November 2015 until November 2017) from 20 oncology centers around Greece. Results In total, 51 (25.5%) patients underwent radical surgical resection of PAC, and 40 (78.4%) of them received adjuvant and 1 (2.0%) neoadjuvant chemotherapy. The median time to recurrence was 7.9 months, and median overall survival (OS), 20.2 months. First-line chemotherapy was administered to 193 (96.5%) patients. The majority of patients were treated with the combination of nab-paclitaxel-gemcitabine (NPG), 5-fluorouracil, leucovorin, irinotecan, oxaliplatin (FOLFIRINOX), or gemcitabine monotherapy. Of them, 39.5% responded to the treatment. Median OS and PFS were 14.1 months and 7.0 months, respectively. Second-line treatment was administered to 112 patients. The majority received NPG, FOLFIRINOX/capecitabine, oxaliplatin, irinotecan (CAPOXIRI), or 5-fluorouracil, leucovorin, oxaliplatin (FOLFOX)/capecitabine, oxaliplatin (CAPOX). Median OS with second-line treatment was 8.6 months, and median PFS, 5.5 months. The most common chemotherapy sequences were NPG as first-line followed by FOLFIRINOX/CAPOXIRI as second-line, NPG followed by FOLFOX/CAPOX, NPG followed by other regimens, and FOLFIRINOX/CAPOXIRI followed by NPG. Conclusion This study described the significant improvement in prognosis of PAC patients receiving palliative chemotherapy and the relatively high rate of receipt of second-line chemotherapy, according to real-world data. However, due to the nonrandomized nature of the study, any comparison between different chemotherapy regimens should be regarded with caution.
Collapse
|
33
|
Patient-Reported Outcome Measures in Pancreatic Cancer Receiving Radiotherapy. Cancers (Basel) 2020; 12:cancers12092487. [PMID: 32887363 PMCID: PMC7563649 DOI: 10.3390/cancers12092487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/31/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Cancer therapies should improve patient survival or at least improve the quality of their life as they receive treatment for their disease. This is particularly important in pancreatic cancer, where current treatments often have to balance between limiting tumor growth and minimizing patient toxicity. There has been an increasing appreciation among physicians to capture the patient’s voice using tools called patient-reported outcome measures (PROM). In this article, we describe the available PROMs and their relative strengths and weaknesses to help oncologists make sense of this rapidly growing field. Finally, we present a decision-making tool that can help researchers and clinicians select the ideal PROM that fits their needs. Abstract Pancreatic cancer and its treatment often dramatically impact patients’ quality of life (QoL). Given this, as well as increased focus on QoL measures in clinical oncology, there has been a rise in the number of instruments that measure patient-reported outcomes (PROs). In this review, we describe the landscape of different PRO instruments pertaining to pancreatic cancer, with specific emphasis on PRO findings related to pancreatic cancer patients receiving radiotherapy (RT). Twenty-five of the most commonly utilized PROs are compared in detail. Notably, most of the PRO tools discussed are not specific to pancreatic cancer but are generic and have been used in various malignancies. Published findings concerning PROs in pancreatic cancer involving RT are also extracted and summarized. Among the measures used, the European Organization for Research and Treatment Cancer QLQ-C30 was the most commonly utilized. We recommend a careful selection of PRO measures in clinical pancreatic cancer research and care and encourage the use of a combination of symptom-specific and global QoL tools to more fully capture patients’ perspectives.
Collapse
|
34
|
Pietrantonio F, Morano F, Niger M, Corallo S, Antista M, Raimondi A, Prisciandaro M, Pagani F, Prinzi N, Nichetti F, Randon G, Torchio M, Corti F, Ambrosini M, Palermo F, Palazzo M, Biamonte L, Platania M, Sposito C, Cosimelli M, Mazzaferro V, Pusceddu S, Cremolini C, de Braud F, Di Bartolomeo M. Systemic Treatment of Patients With Gastrointestinal Cancers During the COVID-19 Outbreak: COVID-19-adapted Recommendations of the National Cancer Institute of Milan. Clin Colorectal Cancer 2020; 19:156-164. [PMID: 32513593 PMCID: PMC7245227 DOI: 10.1016/j.clcc.2020.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/12/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023]
Abstract
The current severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak poses a major challenge in the treatment decision-making of patients with cancer, who may be at higher risk of developing a severe and deadly SARS-CoV-2 infection compared with the general population. The health care emergency is forcing the reshaping of the daily assessment between risks and benefits expected from the administration of immune-suppressive and potentially toxic treatments. To guide our clinical decisions at the National Cancer Institute of Milan (Lombardy region, the epicenter of the outbreak in Italy), we formulated Coronavirus-adapted institutional recommendations for the systemic treatment of patients with gastrointestinal cancers. Here, we describe how our daily clinical practice has changed due to the pandemic outbreak, with the aim of providing useful suggestions for physicians that are facing the same challenges worldwide.
Collapse
Affiliation(s)
- Filippo Pietrantonio
- Oncology and Hemato-oncology Department, University of Milan, Milan, Italy; Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Salvatore Corallo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Antista
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Raimondi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Prisciandaro
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Pagani
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Natalie Prinzi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanni Randon
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Martina Torchio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Margherita Ambrosini
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Palermo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Palazzo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lavinia Biamonte
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco Platania
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carlo Sposito
- Oncology and Hemato-oncology Department, University of Milan, Milan, Italy; Hepato-biliary-pancreatic Surgery and Liver Transplantation Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maurizio Cosimelli
- Colorectal Cancer Surgery Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Vincenzo Mazzaferro
- Oncology and Hemato-oncology Department, University of Milan, Milan, Italy; Hepato-biliary-pancreatic Surgery and Liver Transplantation Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara Cremolini
- Department of Oncology, University Hospital of Pisa, Pisa, Italy; Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Filippo de Braud
- Oncology and Hemato-oncology Department, University of Milan, Milan, Italy; Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
35
|
Di Franco G, Usai A, Funel N, Palmeri M, Montesanti IER, Bianchini M, Gianardi D, Furbetta N, Guadagni S, Vasile E, Falcone A, Pollina LE, Raffa V, Morelli L. Use of zebrafish embryos as avatar of patients with pancreatic cancer: A new xenotransplantation model towards personalized medicine. World J Gastroenterol 2020; 26:2792-2809. [PMID: 32550755 PMCID: PMC7284182 DOI: 10.3748/wjg.v26.i21.2792] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/27/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The response to chemotherapy treatment of patients with pancreatic ductal adenocarcinoma (PDAC) is difficult to predict and the identification of patients who most likely will benefit from aggressive chemotherapy approaches is crucial. The concept of personalized medicine has emerged in the last years with the objective to tailor the medical treatment to the individual characteristics of each patient, and particularly to the tumor biology of each patient. The need for in-vivo xenotransplantation models for cancer patients has increased exponentially, and for this reason zebrafish avatars have gained popularity. Preliminary studies were conducted also with PDAC tissue.
AIM To develop a simple, not expensive, diffusible zebrafish embryo model as avatar for patients affected by PDAC.
METHODS Tumor tissue was taken from the surgical specimen by the histopathologist. After its fragmentation into small pieces, they are stained with CM-Dil. Small pieces of stained tissue were transplanted into the yolk of wt AB zebrafish embryos with a glass capillary needle. Embryos were incubated at 35 °C in E3 medium supplemented with 1% Pen/Strep in the presence or absence of drugs for the following days in respect of the treatment plan (Gemcitabine; Gemcitabine and Oxaliplatin; Gemcitabine and nab-Paclitaxel; 5-Fluorouracil and Folinic acid and Oxaliplatin and Irinotecan). The response of zebrafish xenografts to the chemotherapy options has been analyzed by monitoring the fluorescent stained area at 2 h post injection (hpi), 1 d and 2 d post injection (dpi). In each time point, the mean size of the stained area was measured by ImageJ and it was normalized with respect to the 1 dpi time point mean relative tumor area (RTA). We evaluated the effect of the chemotherapy exposition comparing the mean RTA of each treated subgroup and the control group and evaluating the percentage reduction of the mean RTA by comparing each treated subgroup with the control group.
RESULTS Between July 2018 and October 2019, a total of 15 patients with pancreatic cancer were prospectively enrolled. In all cases, it was possible to take a fragment of the tumor from the surgical specimen for the xenotransplantation in the zebrafish embryos. The histological examination confirmed the presence of a PDAC in all cases. In absence of chemotherapy (control group), over time the Dil-stained area showed a statistically significant increase in all cases. A statistically significant reduction of the mean RTA in the treated subgroups for at least one chemotherapy scheme was reported in 6/15 (40%) cases. The analysis of the percentage reduction of the RTA in treated subgroups in comparison to the control group revealed the presence of a linear relationship in each subgroup between the percentage reduction of the RTA and the number of cases reporting each percentage threshold considered for the analysis.
CONCLUSION Our model seems to be effective for the xenotransplantation of PDAC tissue and evaluation of the effect of each chemotherapy scheme on the xenotransplanted tumor tissue.
Collapse
Affiliation(s)
- Gregorio Di Franco
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Alice Usai
- Department of Biology, University of Pisa, Pisa 56127, Italy
| | - Niccola Funel
- Division of Surgical Pathology, Department of Laboratory of Medicine, Hospital-University of Pisa, Pisa 56124, Italy
| | - Matteo Palmeri
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | | | - Matteo Bianchini
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Desirée Gianardi
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Niccolò Furbetta
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Simone Guadagni
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Enrico Vasile
- Division of Medical Oncology, Pisa University Hospital, Pisa 56124, Italy
| | - Alfredo Falcone
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Luca Emanuele Pollina
- Division of Surgical Pathology, Department of Laboratory of Medicine, Hospital-University of Pisa, Pisa 56124, Italy
| | - Vittoria Raffa
- Department of Biology, University of Pisa, Pisa 56127, Italy
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
- EndoCAS (Center for Computer Assisted Surgery), University of Pisa, Pisa 56124, Italy
| |
Collapse
|
36
|
Vienot A, Chevalier H, Bolognini C, Gherga E, Klajer E, Meurisse A, Jary M, Kim S, d’Engremont C, Nguyen T, Calcagno F, Almotlak H, Fein F, Nasri M, Abdeljaoued S, Turpin A, Borg C, Vernerey D. FOLFOXIRI vs FOLFIRINOX as first-line chemotherapy in patients with advanced pancreatic cancer: A population-based cohort study. World J Gastrointest Oncol 2020; 12:332-346. [PMID: 32206183 PMCID: PMC7081111 DOI: 10.4251/wjgo.v12.i3.332] [Citation(s) in RCA: 3] [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: 08/24/2019] [Revised: 12/26/2019] [Accepted: 01/14/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND FOLFIRINOX regimen is the first-line reference chemotherapy (L1) in advanced pancreatic ductal adenocarcinoma (aPDAC). FOLFOXIRI, a schedule with a lower dose of irinotecan and no bolus 5-fluorouracil, has demonstrated efficacy and feasibility in colorectal cancer.
AIM To investigate the potential clinical value of FOLFOXIRI in patients with aPDAC in routine clinical practice.
METHODS Analyses were derived from all consecutive aPDAC patients treated in L1 between January 2011 and December 2017 in two French institutions, with either FOLFOXIRI (n = 165) or FOLFIRINOX (n = 124) regimens. FOLFOXIRI consisted of irinotecan (165 mg/m2), oxaliplatin (85 mg/m2), leucovorin (200 mg/m2) and 5-fluorouracil (3200 mg/m2 as a 48-h continuous infusion) every 2 wk. Ninety-six pairs of patients were selected through propensity score matching, and clinical outcomes of the two treatment regimens were compared.
RESULTS Median overall survival was 11.1 mo in the FOLFOXIRI and 11.6 mo in the FOLFIRINOX cohorts, respectively. After propensity score matching, survival rates remained similar between the two regimens in terms of overall survival (hazard ratio = 1.22; P = 0.219) and progression-free survival (hazard ratio = 1.27; P = 0.120). The objective response rate was 37.1% in the FOLFOXIRI group vs 47.8% in the FOLFIRINOX group (P = 0.187). Grade 3/4 toxicities occurred in 28.7% of patients in the FOLFOXIRI cohort vs 19.5% in the FOLFIRINOX cohort (P = 0.079). FOLFOXIRI was associated with a higher incidence of grade 3/4 digestive adverse events. Hematopoietic growth factors were used after each chemotherapy cycle and the low hematological toxicity rates were below 5% with both regimens.
CONCLUSION FOLFOXIRI is feasible in L1 in patients with aPDAC but does not confer any therapeutic benefit as compared with FOLFIRINOX. The low hematological toxicity rates strengthened the relevance of primary prophylaxis with hematopoietic growth factors.
Collapse
Affiliation(s)
- Angélique Vienot
- Department of Medical Oncology, Besançon University Hospital, Besançon F-25030, France
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- INSERM CIC-1431, Clinical Investigation Center in Biotherapy, Besançon University Hospital, Besançon F-25030, France
| | - Hortense Chevalier
- Department of Medical Oncology, Lille University Hospital, Lille F-59000, France
| | - Clément Bolognini
- Department of Medical Oncology, Besançon University Hospital, Besançon F-25030, France
| | - Elisabeta Gherga
- Department of Medical Oncology, Nord Franche-Comté Hospital, Montbéliard F-25020, France
| | - Elodie Klajer
- Department of Medical Oncology, Besançon University Hospital, Besançon F-25030, France
| | - Aurélia Meurisse
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- Methodological and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon F-25030, France
| | - Marine Jary
- Department of Medical Oncology, Besançon University Hospital, Besançon F-25030, France
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- INSERM CIC-1431, Clinical Investigation Center in Biotherapy, Besançon University Hospital, Besançon F-25030, France
| | - Stefano Kim
- Department of Medical Oncology, Besançon University Hospital, Besançon F-25030, France
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- INSERM CIC-1431, Clinical Investigation Center in Biotherapy, Besançon University Hospital, Besançon F-25030, France
| | | | - Thierry Nguyen
- Department of Medical Oncology, Besançon University Hospital, Besançon F-25030, France
| | - Fabien Calcagno
- Department of Medical Oncology, Besançon University Hospital, Besançon F-25030, France
| | - Hamadi Almotlak
- Department of Medical Oncology, Besançon University Hospital, Besançon F-25030, France
| | - Francine Fein
- Department of Gastroenterology, Besançon University Hospital, Besançon F-25030, France
| | - Meher Nasri
- Department of Medical Oncology, Nord Franche-Comté Hospital, Montbéliard F-25020, France
| | - Syrine Abdeljaoued
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
| | - Anthony Turpin
- Department of Medical Oncology, Lille University Hospital, Lille F-59000, France
| | - Christophe Borg
- Department of Medical Oncology, Besançon University Hospital, Besançon F-25030, France
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- INSERM CIC-1431, Clinical Investigation Center in Biotherapy, Besançon University Hospital, Besançon F-25030, France
| | - Dewi Vernerey
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- Methodological and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon F-25030, France
| |
Collapse
|
37
|
Moving the Target on the Optimal Adjuvant Strategy for Resected Pancreatic Cancers: A Systematic Review with Meta-Analysis. Cancers (Basel) 2020; 12:cancers12030534. [PMID: 32110977 PMCID: PMC7139837 DOI: 10.3390/cancers12030534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023] Open
Abstract
Combination regimens have shown superiority over single agents in the adjuvant treatment of resected pancreatic cancer (PC), but there are no data supporting definition of the best regimen. This work aimed to compare the efficacy and safety of mFOLFIRINOX, gemcitabine+capecitabine, and gemcitabine+nab/paclitaxel in PC patients. A meta-analysis was performed for direct comparison between trials comparing combination regimens and gemcitabine monotherapy. Subsequently, an indirect comparison was made between trials investigating the efficacy and safety of mFOLFIRINOX, gemcitabine+capecitabine, and gemcitabine+nab/paclitaxel because of the same control arm (gemcitabine). A total of three studies met the selection criteria and were included in our indirect comparison. Indirect comparisons for efficacy outcomes showed a benefit in terms of DFS (disease-free survival)/EFS (event-free survival)/RFS (relapse-free survival) for both mFOLFIRINOX versus gemcitabine+capecitabine (HR 0.69, 95% CI 0.52–0.91) and versus gemcitabine+nab/paclitaxel (HR 0.67, 95% CI 0.50–0.90). No significant advantage was registered for OS (overall survival). Indirect comparisons for safety showed an increase in terms of G3-5 AEs (with the exception of neutropenia) for mFOLFIRINOX versus gemcitabine+capecitabine (RR 1.24, 95% CI 1.03–1.50), while no significant differences were observed versus gemcitabine+nab/paclitaxel. According to our results, mFOLFIRINOX is feasible and manageable and could represent a first option for fit PC resected patients.
Collapse
|
38
|
Ye J, Mills BN, Zhao T, Han BJ, Murphy JD, Patel AP, Johnston CJ, Lord EM, Belt BA, Linehan DC, Gerber SA. Assessing the Magnitude of Immunogenic Cell Death Following Chemotherapy and Irradiation Reveals a New Strategy to Treat Pancreatic Cancer. Cancer Immunol Res 2020; 8:94-107. [PMID: 31719057 PMCID: PMC6946873 DOI: 10.1158/2326-6066.cir-19-0373] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/18/2019] [Accepted: 11/07/2019] [Indexed: 12/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to have a dismal prognosis, in part, due to ineffective treatment strategies. The efficacy of some chemotherapies and especially radiotherapy is mediated partially by the immune system. Therefore, we hypothesized that profiling the immune response following chemotherapy and/or irradiation can be used as a readout for treatment efficacy but also to help identify optimal therapeutic schedules for PDAC. Using murine models of PDAC, we demonstrated that concurrent administration of stereotactic body radiotherapy (SBRT) and a modified dose of FOLFIRINOX (mFX) resulted in superior tumor control when compared with single or sequential treatment groups. Importantly, this combined treatment schedule enhanced the magnitude of immunogenic cell death, which in turn amplified tumor antigen presentation by dendritic cells and intratumoral CD8+ T-cell infiltration. Concurrent therapy also resulted in systemic immunity contributing to the control of established metastases. These findings provide a rationale for pursuing concurrent treatment schedules of SBRT with mFX in PDAC.
Collapse
Affiliation(s)
- Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Bradley N Mills
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Tony Zhao
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Booyeon J Han
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Joseph D Murphy
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Ankit P Patel
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Carl J Johnston
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Edith M Lord
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Brian A Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - David C Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Scott A Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, New York.
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
39
|
The efficacy and toxicity of chemotherapy in the elderly with advanced pancreatic cancer. Pancreatology 2020; 20:95-100. [PMID: 31786057 DOI: 10.1016/j.pan.2019.11.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVES FOLFIRINOX (FFX) or abraxane plus gemcitabine (AG)-based chemotherapy is used widely as firstline treatment for patients with pancreatic cancer. However, their use in the elderly is discouraged because of adverse events. More clinical data about the therapeutic response and tolerability to FFX or AG in elderly patents (over 70 years old) are required. METHODS Patients with advanced pancreatic cancer (n = 203; 131 metastatic pancreatic cancer patients (MPC) and 72 locally advanced pancreatic cancer patients (LAPC)) were treated using modified-FFX (mFFX) or AG and mFFX sequentially. The patients were grouped according to their age, patients below 70 years old and patients above 70 years old. The objective response rate (ORR), disease control rate (DCR), progression free survival (PFS), overall survival (OS) and adverse events were compared between the groups. RESULTS The ORRs in the elderly and in patients below 70 were similar (30.0% versus 32.3%). The median OS and PFS were also similar between the groups (mOS 13.3 m vs 12.7 m, p = 0.729, HR 0.874 (95% CI 0.5310 to 1.438); mPFS mPFS 10.6 m vs 10.3 m, p = 0.363, HR 0.800 (95% CI 0.4954 to 1.293)). However, the elderly patients suffered a higher incidence of severe adverse events (50% vs. 28.3%). CONCLUSIONS These data could provide guidance for chemotherapy use in elderly patients with advanced pancreatic cancer. Age did not affect treatment outcome; however, supportive treatment is very important for elderly patients receiving chemotherapy.
Collapse
|
40
|
Law HCH, Lagundžin D, Clement EJ, Qiao F, Wagner ZS, Krieger KL, Costanzo-Garvey D, Caffrey TC, Grem JL, DiMaio DJ, Grandgenett PM, Cook LM, Fisher KW, Yu F, Hollingsworth MA, Woods NT. The Proteomic Landscape of Pancreatic Ductal Adenocarcinoma Liver Metastases Identifies Molecular Subtypes and Associations with Clinical Response. Clin Cancer Res 2019; 26:1065-1076. [PMID: 31848187 DOI: 10.1158/1078-0432.ccr-19-1496] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 10/19/2019] [Accepted: 12/11/2019] [Indexed: 12/16/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic disease that can be separated into distinct subtypes based on molecular signatures. Identifying PDAC subtype-specific therapeutic vulnerabilities is necessary to develop precision medicine approaches to treat PDAC. EXPERIMENTAL DESIGN A total of 56 PDAC liver metastases were obtained from the UNMC Rapid Autopsy Program and analyzed with quantitative proteomics. PDAC subtypes were identified by principal component analysis based on protein expression profiling. Proteomic subtypes were further characterized by the associated clinical information, including but not limited to survival analysis, drug treatment response, and smoking and drinking status. RESULTS Over 3,960 proteins were identified and used to delineate four distinct PDAC microenvironment subtypes: (i) metabolic; (ii) progenitor-like; (iii) proliferative; and (iv) inflammatory. PDAC risk factors of alcohol and tobacco consumption correlate with subtype classifications. Enhanced survival is observed in FOLFIRINOX treated metabolic and progenitor-like subtypes compared with the proliferative and inflammatory subtypes. In addition, TYMP, PDCD6IP, ERAP1, and STMN showed significant association with patient survival in a subtype-specific manner. Gemcitabine-induced alterations in the proteome identify proteins, such as serine hydroxymethyltransferase 1, associated with drug resistance. CONCLUSIONS These data demonstrate that proteomic analysis of clinical PDAC liver metastases can identify molecular signatures unique to disease subtypes and point to opportunities for therapeutic development to improve the treatment of PDAC.
Collapse
Affiliation(s)
- Henry C-H Law
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Dragana Lagundžin
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Emalie J Clement
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Fangfang Qiao
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Zachary S Wagner
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kimiko L Krieger
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Diane Costanzo-Garvey
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha Nebraska
| | - Thomas C Caffrey
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jean L Grem
- Internal Medicine, Division of Hematology Oncology, University of Nebraska Medical Center, Omaha Nebraska
| | - Dominick J DiMaio
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha Nebraska
| | - Paul M Grandgenett
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Leah M Cook
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha Nebraska
| | - Kurt W Fisher
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha Nebraska
| | - Fang Yu
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha Nebraska
| | - Michael A Hollingsworth
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Nicholas T Woods
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska.
| |
Collapse
|
41
|
Kim YI, Song KB, Lee YJ, Park KM, Hwang DW, Lee JH, Shin SH, Kwon JW, Ro JS, Kim SC. Management of isolated recurrence after surgery for pancreatic adenocarcinoma. Br J Surg 2019; 106:898-909. [PMID: 31162655 DOI: 10.1002/bjs.11144] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/10/2018] [Accepted: 01/25/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Recurrence of pancreatic cancer after primary pancreatectomy occurs in the vast majority of patients. The role of surgical treatment for recurrent pancreatic cancer is not well established. METHODS Patients who underwent primary pancreatectomy with curative intent from 2000 to 2014 at a single large-volume centre were evaluated retrospectively. CT or PET was used to select patients with an isolated recurrence. The clinicopathological features and survival outcomes were compared according to treatment modalities. RESULTS Of the 1610 patients with pancreatic cancer who underwent resection, 1346 (83·6 per cent) were diagnosed with recurrent pancreatic cancer. Recurrence was locoregional in 366 patients (27·2 per cent), distant multifocal in 251 (18·6 per cent), distant isolated in 188 (14·0 per cent), locoregional plus distant in 153 (11·4 per cent) and peritoneal seeding in 388 (28·8 per cent). Of the 1346 patients with recurrence, 197 (14·6 per cent) had isolated recurrence; of these, 48 (24·4 per cent of all isolated recurrences; 3·6 per cent of all recurrences) underwent resection. Median survival of the 197 patients after diagnosis of isolated recurrence was 14·7 months; it was longer in patients who underwent surgical resection than among those treated non-surgically (23·5 versus 12·0 months; P = 0·014). Multivariable analysis showed that chemotherapy and resection for recurrence were associated with better prognosis. Median survival after recurrence was longest in the 23 patients with isolated pulmonary recurrence (33·3 months). Survival after recurrence was better in patients who underwent resection of isolated recurrence in the remnant pancreas (median 28·0 versus 12·0 months, P = 0·010) and lung (median 36·5 versus 9·5 months; P = 0·010) than in those who did not undergo resection. CONCLUSION Surgical resection may be considered an option for treatment of patients with isolated recurrent pancreatic cancer.
Collapse
Affiliation(s)
- Y I Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, South Korea
| | - K B Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, South Korea
| | - Y-J Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, South Korea
| | - K-M Park
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, South Korea
| | - D W Hwang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, South Korea
| | - J H Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, South Korea
| | - S H Shin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, South Korea
| | - J W Kwon
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, South Korea
| | - J-S Ro
- Clinical Preventive Medicine Centre, Seoul National University Bundang Hospital, Bundang-gu, South Korea
| | - S C Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, South Korea
| |
Collapse
|
42
|
Hyde AJ, Nassabein R, AlShareef A, Armstrong D, Babak S, Berry S, Bossé D, Chen E, Colwell B, Essery C, Goel R, Goodwin R, Gray S, Hammad N, Jeyakuymar A, Jonker D, Karanicolas P, Lamond N, Letourneau R, Michael J, Patil N, Powell E, Ramjeesingh R, Saliba W, Singh R, Snow S, Stuckless T, Tadros S, Tehfé M, Thana M, Thirlwell M, Vickers M, Virik K, Welch S, Asmis T. Eastern Canadian Gastrointestinal Cancer Consensus Conference 2018. Curr Oncol 2019; 26:e665-e681. [PMID: 31708660 PMCID: PMC6821113 DOI: 10.3747/co.26.5193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The annual Eastern Canadian Gastrointestinal Cancer Consensus Conference was held in Halifax, Nova Scotia, 20-22 September 2018. Experts in radiation oncology, medical oncology, surgical oncology, and pathology who are involved in the management of patients with gastrointestinal malignancies participated in presentations and discussion sessions for the purpose of developing the recommendations presented here. This consensus statement addresses multiple topics in the management of pancreatic cancer, pancreatic neuroendocrine tumours, hepatocellular cancer, and rectal and colon cancer, including ■ surgical management of pancreatic adenocarcinoma,■ adjuvant and metastatic systemic therapy options in pancreatic adenocarcinoma,■ the role of radiotherapy in the management of pancreatic adenocarcinoma,■ systemic therapy in pancreatic neuroendocrine tumours,■ updates in systemic therapy for patients with advanced hepatocellular carcinoma,■ optimum duration of adjuvant systemic therapy for colorectal cancer, and■ sequence of therapy in oligometastatic colorectal cancer.
Collapse
Affiliation(s)
- A J Hyde
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - R Nassabein
- Quebec-McGill University Health Centre, Montreal (Thirlwell); Centre Hospitalier de l'Université de Montréal, Montreal (Letourneau, Nassabein, Tehfé)
| | - A AlShareef
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - D Armstrong
- Newfoundland and Labrador-Dr. H. Bliss Murphy Cancer Centre, St. John's (Armstrong, Powell, Stuckless)
| | - S Babak
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - S Berry
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - D Bossé
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - E Chen
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - B Colwell
- Nova Scotia-Queen Elizabeth ii Health Sciences Centre, Dalhousie University, Halifax (Colwell, Jeyakumar, Lamond, Patil, Ramjeesingh, Singh, Saliba, Snow, Thana)
| | - C Essery
- New Brunswick-Saint John Regional Hospital, Saint John (Gray, Michael)
| | - R Goel
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - R Goodwin
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - S Gray
- British Columbia-Penticton Regional Hospital, Penticton (Essery)
| | - N Hammad
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - A Jeyakuymar
- Nova Scotia-Queen Elizabeth ii Health Sciences Centre, Dalhousie University, Halifax (Colwell, Jeyakumar, Lamond, Patil, Ramjeesingh, Singh, Saliba, Snow, Thana)
| | - D Jonker
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - P Karanicolas
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - N Lamond
- Nova Scotia-Queen Elizabeth ii Health Sciences Centre, Dalhousie University, Halifax (Colwell, Jeyakumar, Lamond, Patil, Ramjeesingh, Singh, Saliba, Snow, Thana)
| | - R Letourneau
- Quebec-McGill University Health Centre, Montreal (Thirlwell); Centre Hospitalier de l'Université de Montréal, Montreal (Letourneau, Nassabein, Tehfé)
| | - J Michael
- British Columbia-Penticton Regional Hospital, Penticton (Essery)
| | - N Patil
- Nova Scotia-Queen Elizabeth ii Health Sciences Centre, Dalhousie University, Halifax (Colwell, Jeyakumar, Lamond, Patil, Ramjeesingh, Singh, Saliba, Snow, Thana)
| | - E Powell
- Newfoundland and Labrador-Dr. H. Bliss Murphy Cancer Centre, St. John's (Armstrong, Powell, Stuckless)
| | - R Ramjeesingh
- Nova Scotia-Queen Elizabeth ii Health Sciences Centre, Dalhousie University, Halifax (Colwell, Jeyakumar, Lamond, Patil, Ramjeesingh, Singh, Saliba, Snow, Thana)
| | - W Saliba
- Nova Scotia-Queen Elizabeth ii Health Sciences Centre, Dalhousie University, Halifax (Colwell, Jeyakumar, Lamond, Patil, Ramjeesingh, Singh, Saliba, Snow, Thana)
| | - R Singh
- Nova Scotia-Queen Elizabeth ii Health Sciences Centre, Dalhousie University, Halifax (Colwell, Jeyakumar, Lamond, Patil, Ramjeesingh, Singh, Saliba, Snow, Thana)
| | - S Snow
- Nova Scotia-Queen Elizabeth ii Health Sciences Centre, Dalhousie University, Halifax (Colwell, Jeyakumar, Lamond, Patil, Ramjeesingh, Singh, Saliba, Snow, Thana)
| | - T Stuckless
- Newfoundland and Labrador-Dr. H. Bliss Murphy Cancer Centre, St. John's (Armstrong, Powell, Stuckless)
| | - S Tadros
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - M Tehfé
- Quebec-McGill University Health Centre, Montreal (Thirlwell); Centre Hospitalier de l'Université de Montréal, Montreal (Letourneau, Nassabein, Tehfé)
| | - M Thana
- Nova Scotia-Queen Elizabeth ii Health Sciences Centre, Dalhousie University, Halifax (Colwell, Jeyakumar, Lamond, Patil, Ramjeesingh, Singh, Saliba, Snow, Thana)
| | - M Thirlwell
- Quebec-McGill University Health Centre, Montreal (Thirlwell); Centre Hospitalier de l'Université de Montréal, Montreal (Letourneau, Nassabein, Tehfé)
| | - M Vickers
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - K Virik
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - S Welch
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| | - T Asmis
- Ontario-The Ottawa Hospital Cancer Centre, Ottawa (AlShareef, Asmis, Bossé, Goel, Goodwin, Hyde, Jonker, Tadros, Vickers); Queen's University and Cancer Centre of Southeastern Ontario, Kingston (Hammad, Virik); Princess Margaret Cancer Centre, Toronto (Chen); Markham Stouffville Hospital, Markham (Babak); Sunnybrook Odette Cancer Centre, University of Toronto, Toronto (Berry, Karanicolas); London Health Sciences Centre, London (Welch)
| |
Collapse
|
43
|
Costa JG, de Jesus VHF, Camandaroba MPG, Dettino ALA. Characteristics and survival of older patients with metastatic pancreatic cancer: a retrospective analysis of the AC Camargo Cancer Center experience. Ther Adv Med Oncol 2019; 11:1758835919874650. [PMID: 31534492 PMCID: PMC6737872 DOI: 10.1177/1758835919874650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 07/25/2019] [Indexed: 12/28/2022] Open
Abstract
Background: Advanced age is the most important risk factor for pancreatic cancer and about half of patients are diagnosed with metastatic disease. In the first-line setting, multidrug chemotherapy regimens were shown to be more effective than gemcitabine alone. However, the older population was under-represented in randomized clinical trials. We aimed to describe the clinical profile of older patients with metastatic pancreatic cancer and their survival outcomes. Materials and methods: This was a retrospective, unicentric study that included patients diagnosed with metastatic pancreatic cancer (non-neuroendocrine), aged 65 years and over. Results: The study population comprised 196 patients. The median age was 73 years; 67% of these patients presented Eastern Cooperative Oncology Group performance status (ECOG) ⩽ 1 and the median Charlson Comorbidity score was 10. Chemotherapy was given to 89% of the patients. The most frequently used chemotherapy regimens were gemcitabine (44%), 5-fluorouracil and oxaliplatin [FOLFOX; 26%], and 5-fluorouracil, oxaliplatin and irinotecan (FOLFIRINOX; 20%). Patients treated with FOLFIRINOX were younger and they presented better performance status. After a median follow up of 19.8 months, the median overall survival (OS) was of 7.2 months and the median time to first-line-treatment failure was 4.6 months. Among patients treated with chemotherapy, the median OS was highest for those treated with FOLFIRINOX (13.8 months), as compared with FOLFOX (7.0 months) or gemcitabine (6.7 months); p = 0.004. Nonetheless, treatment with FOLFIRINOX was associated with increased risk of severe toxicity (p = 0.008). Conclusion: Older patients with metastatic pancreatic cancer benefit from palliative chemotherapy, and FOLFIRINOX is a therapeutic option in rigorously selected older patients.
Collapse
Affiliation(s)
| | - Victor Hugo Fonseca de Jesus
- Medical Oncology Department AC Camargo Cancer Center Rua Prof. Antônio Prudente, 211 - 01509-010, São Paulo, Brazil
| | | | | |
Collapse
|
44
|
Pusceddu S, Ghidini M, Torchio M, Corti F, Tomasello G, Niger M, Prinzi N, Nichetti F, Coinu A, Di Bartolomeo M, Cabiddu M, Passalacqua R, de Braud F, Petrelli F. Comparative Effectiveness of Gemcitabine plus Nab-Paclitaxel and FOLFIRINOX in the First-Line Setting of Metastatic Pancreatic Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2019; 11:E484. [PMID: 30959763 PMCID: PMC6520876 DOI: 10.3390/cancers11040484] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/26/2019] [Accepted: 03/28/2019] [Indexed: 12/18/2022] Open
Abstract
Gemcitabine and nab-paclitaxel (GEM-NAB) and the combination of 5-fluorouracil, oxaliplatin, and irinotecan (FOLFIRINOX) are valid first-line options for advanced or metastatic pancreatic cancer (mPC). However, no randomized trials comparing the two schemes have been performed. This meta-analysis aims to compare GEM-NAB and FOLFIRINOX in terms of safety and effectiveness, taking into account data from real-life studies on mPC. We systematically searched PubMed, EMBASE and Cochrane library up to November 2018 to identify retrospective or cohort studies on mPC comparing GEM-NAB and FOLFIRINOX. We included 16 retrospective studies, including 3813 patients (2123 treated with GEM-NAB and 1690 treated with FOLFIRINOX). Despite a median weighted overall survival (OS) difference in favor of FOLFIRINOX (mean difference: 1.15, 95% confidence interval CI 0.08⁻2.22, p = 0.03), in whole population OS was similar (hazard ratio (HR = 0.99, 95% CI 0.84⁻1.16; p = 0.9). PFS was also not different between the two arms (HR = 0.88, 95% CI 0.71⁻1.1; p = 0.26). The overall response rate was similar (25 vs. 24% with GEM-NAB and FOLFIRINOX). Among grade 3⁻4 toxicities, neutropenia, febrile neutropenia, and nausea were lower with GEM-NAB, while neurotoxicity and anemia were lower with FOLFIRINOX. In conclusion, despite a numerically longer median OS with FOLFIRINOX as compared to GEM-NAB, the overall risk of death and progression were similar. Their toxicity was different with less nausea, neutropenia, and febrile neutropenia with GEM-NAB, as compared to less neurotoxicity and anemia with FOLFIRINOX. Therefore, analysis of non-randomized "real world" studies to date has not provided evidence of a major benefit of one regimen over the other.
Collapse
Affiliation(s)
- Sara Pusceddu
- Medical Oncology Department, Fondazione IRCSS Istituto Nazionale Tumori di Milano, 20133 Milan, Italy.
| | - Michele Ghidini
- Medical Oncology Department, ASST Cremona, 26100 Cremona, Italy.
| | - Martina Torchio
- Medical Oncology Department, Fondazione IRCSS Istituto Nazionale Tumori di Milano, 20133 Milan, Italy.
| | - Francesca Corti
- Medical Oncology Department, Fondazione IRCSS Istituto Nazionale Tumori di Milano, 20133 Milan, Italy.
| | | | - Monica Niger
- Medical Oncology Department, Fondazione IRCSS Istituto Nazionale Tumori di Milano, 20133 Milan, Italy.
| | - Natalie Prinzi
- Medical Oncology Department, Fondazione IRCSS Istituto Nazionale Tumori di Milano, 20133 Milan, Italy.
| | - Federico Nichetti
- Medical Oncology Department, Fondazione IRCSS Istituto Nazionale Tumori di Milano, 20133 Milan, Italy.
| | - Andrea Coinu
- Medical Oncology Department, Ospedale San Francesco, ASSL Nuoro, 08100 Nuoro, Italy.
| | - Maria Di Bartolomeo
- Medical Oncology Department, Fondazione IRCSS Istituto Nazionale Tumori di Milano, 20133 Milan, Italy.
| | - Mary Cabiddu
- Medical Oncology and Hemato-Oncology Department, ASST Bergamo Ovest, 24047 Bergamo, Italy.
| | | | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCSS Istituto Nazionale Tumori di Milano, 20133 Milan, Italy.
- Department, University of Milan, 20122 Milan, Italy.
| | - Fausto Petrelli
- Medical Oncology and Hemato-Oncology Department, ASST Bergamo Ovest, 24047 Bergamo, Italy.
| |
Collapse
|
45
|
Sasaki T, Kanata R, Yamada I, Matsuyama M, Ozaka M, Sasahira N. Improvement of Treatment Outcomes for Metastatic Pancreatic Cancer: A Real-world Data Analysis. In Vivo 2019; 33:271-276. [PMID: 30587635 DOI: 10.21873/invivo.11471] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/11/2018] [Accepted: 11/15/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIM FOLFIRINOX (5-fluorouracil, leucovorin, irinotecan, oxaliplatin) and gemcitabine plus nab-paclitaxel therapy have recently been introduced for the treatment of metastatic pancreatic cancer. Herein, overall treatment outcomes of metastatic pancreatic cancer after introduction of FOLFIRINOX and gemcitabine plus nab-paclitaxel therapy were evaluated, in daily practice. PATIENTS AND METHODS Metastatic pancreatic cancer patients (n=321) who started systemic chemotherapy between January 2011 and December 2016 were included and were divided into two groups: group A (2011-2013) and group B (2014-2016). Treatment outcomes were evaluated retrospectively. RESULTS Patient characteristics were similar between the two groups except for the rates of distant lymph node metastasis and peritoneal metastasis. The preferred regimens in groups A and B were gemcitabine monotherapy and gemcitabine plus nab-paclitaxel therapy, respectively. The response rates, median progression-free survival, and median overall survival of groups A and B were 7.8% and 28.4% (p<0.01), 3.1 months and 5.4 months (p<0.01), and 6.7 months and 10.2 months (p<0.01), respectively. CONCLUSION Overall treatment outcomes for metastatic pancreatic cancer were significantly improved after introduction of FOLFIRINOX and gemcitabine plus nab-paclitaxel combination therapy in daily practice.
Collapse
Affiliation(s)
- Takashi Sasaki
- Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Ryo Kanata
- Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Ikuhiro Yamada
- Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Masato Matsuyama
- Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Masato Ozaka
- Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Naoki Sasahira
- Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation of Cancer Research, Tokyo, Japan
| |
Collapse
|
46
|
Eraslan E, Yildiz F, Tufan G, Aslan F, Demirci U, Oksuzoglu OB. First line modified Folfirinox versus gemcitabine for advanced pancreatic cancer: A single institution retrospective experience. JOURNAL OF ONCOLOGICAL SCIENCES 2019. [DOI: 10.1016/j.jons.2019.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
47
|
Ulusakarya A, Teyar N, Karaboué A, Haydar M, Krimi S, Biondani P, Gumus Y, Chebib A, Almohamad W, Morère JF. Patient-tailored FOLFIRINOX as first line treatment of patients with advanced pancreatic adenocarcinoma. Medicine (Baltimore) 2019; 98:e15341. [PMID: 31008993 PMCID: PMC6494255 DOI: 10.1097/md.0000000000015341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
FOLFIRINOX is one of the most effective reference regimens in the 1st line treatment of locally advanced (LA) and metastatic pancreatic cancer (mPC), despite its high toxicity. We evaluated our real-life experience with "patient-tailored intent to treat FOLFIRINOX" in patients with LA or mPC compared to other reports along with the pivotal phase III trial.We analyzed data from all consecutive patients with pancreatic ductal adenocarcinoma treated with dose-modified FOLFIRINOX in 2016 at Paul Brousse University Hospital. Irinotecan was administered whenever initial serum bilirubin was <1.5 × upper limit of normal. Oxaliplatin was stopped for severe sensory neuropathy. Initial dose reductions were made according to patient profile (eg, age, comorbidities) and later due to toxicity. The treatment was continued until surgery or disease progression. Endpoints were time to progression (TTP), overall survival (OS), objective response rate (ORR), and secondary complete resection (R0R1).Thirty-seven patients with unresectable LA or mPC received patient-tailored FOLFIRINOX as 1st line chemotherapy. There were 22 male (59%) and 15 female patients (41%) aged 44 to 81 years with LA (18 patients, 49%) and mPC (19 patients, 51%). They had World Health Organization-performance status of 0 (59%) or 1 (41%). A total of 384 cycles were administered. Median dose intensities (mg/m/w) were 28.9 for oxaliplatin, 56.8 for irinotecan, and 886.2 for 5-fluorouracil. Thirty-four patients were assessed for response; ORR and disease control rates were 47% and 85%, respectively. R0R1 rate was 30%. Median TTP and OS were 9.6 and 14.6 months. LA disease was associated with significantly longer TTP and OS (P < .001).FOLFIRINOX with patient-tailored dose adaptations seems to offer better results in patients with advanced PC. This approach in the neoadjuvant setting results in a macroscopic R0R1 in 61% of patients with initially unresectable disease. It deserves prospective evaluation to further improve outcomes in the management of advanced PC.
Collapse
Affiliation(s)
- Ayhan Ulusakarya
- Assistance Publique-Hôpitaux de Paris, Department of Medical Oncology, Paul Brousse Hospital
- INSERM U935 Campus CNRS, Villejuif
| | - Nahla Teyar
- Assistance Publique-Hôpitaux de Paris, Department of Medical Oncology, Paul Brousse Hospital
| | - Abdoulaye Karaboué
- INSERM U935 Campus CNRS, Villejuif
- Medical Oncology Unit, GHI Le Raincy-Montfermeil, Montfermeil, France
| | - Mazen Haydar
- Assistance Publique-Hôpitaux de Paris, Department of Medical Oncology, Paul Brousse Hospital
| | - Sarra Krimi
- Assistance Publique-Hôpitaux de Paris, Department of Medical Oncology, Paul Brousse Hospital
| | - Pamela Biondani
- Assistance Publique-Hôpitaux de Paris, Department of Medical Oncology, Paul Brousse Hospital
| | - Yusuf Gumus
- Assistance Publique-Hôpitaux de Paris, Department of Medical Oncology, Paul Brousse Hospital
| | - Amale Chebib
- Assistance Publique-Hôpitaux de Paris, Department of Medical Oncology, Paul Brousse Hospital
| | - Wathek Almohamad
- Assistance Publique-Hôpitaux de Paris, Department of Medical Oncology, Paul Brousse Hospital
| | - Jean-François Morère
- Assistance Publique-Hôpitaux de Paris, Department of Medical Oncology, Paul Brousse Hospital
| |
Collapse
|
48
|
Khader S, Thyagarajan A, Sahu RP. Exploring Signaling Pathways and Pancreatic Cancer Treatment Approaches Using Genetic Models. Mini Rev Med Chem 2019; 19:1112-1125. [PMID: 30924420 DOI: 10.2174/1389557519666190327163644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/12/2019] [Accepted: 03/19/2019] [Indexed: 11/22/2022]
Abstract
Despite available treatment options, the overall survival rates of pancreatic cancer patients remain dismal. Multiple counter-regulatory pathways have been identified and shown to be involved in interfering with the efficacy of therapeutic agents. In addition, various known genetic alterations in the cellular signaling pathways have been implicated in affecting the growth and progression of pancreatic cancer. Nevertheless, the significance of other unknown pathways is yet to be explored, which provides the rationale for the intervention of new approaches. Several experimental genetic models have been explored to define the impact of key signaling cascades, and their mechanisms in the pathophysiology as well as treatment approaches of pancreatic cancer. The current review highlights the recent updates, and significance of such genetic models in the therapeutic efficacy of anti-tumor agents including the standard chemotherapeutic agents, natural products, cell signaling inhibitors, immunebased therapies and the combination of these approaches in pancreatic cancer.
Collapse
Affiliation(s)
- Shorooq Khader
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH 45345, United States
| | - Anita Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH 45345, United States
| | - Ravi P Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH 45345, United States
| |
Collapse
|