1
|
Sezgin Y, Karhan O, Aldemir MN, Ürün M, Erçek BM, Urakcı Z, Arvas H, Tunç S, Erdem M, Yerlikaya H, İleri S, Aydın İ, Bicer A, Kömüroğlu AU, Majidova N, Gökçek S, Demir H, Yıldız S, Akbaş S, Özen E, Kahya BU, Sali M, Anık H, Aykut T, Araz M, Alkan A, Özçelik M, Sakin A, Aykan MB, Mehtıyev M, Demir B, Başer MN, Sönmez M, Gültürk İ, Avcı N, Urvay S, Arıcı MÖ, Kalender ME, Yıldırım M, Solmaz AA, Gürbüz M, Ergün Y. Efficacy of gemcitabine plus nab-paclitaxel in second-line treatment of metastatic pancreatic cancer. Sci Rep 2025; 15:11675. [PMID: 40188172 PMCID: PMC11972389 DOI: 10.1038/s41598-025-96157-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Despite numerous studies on second-line therapies in metastatic pancreatic cancer, there is no randomized study evaluating the efficacy of gemcitabine plus nab-paclitaxel as a second-line treatment. This study aims to examine the efficacy of gemcitabine plus nab-paclitaxel in second-line therapy. In this retrospective study, a total of 218 patients from 23 centers were included. The primary endpoint was progression-free survival (PFS), secondary endpoints included overall survival (OS), treatment efficacy based on ECOG performance status (PS), and tumor marker (CEA, CA 19 - 9) levels. In the second-line treatment with gemcitabine plus nab-paclitaxel, the median PFS was 5.1 months (95% CI, 5.6 to 7.1), and the median OS was 8.6 months (95% CI, 7.3 to 10.0). Median PFS was 6.6 months in patients with normal CEA levels compared to 4.4 months in patients with high CEA levels (P = 0.01). Median PFS was 6 months in patients with ECOG PS 0-1 compared to 3.8 months in patients with PS 2 (P < 0.01). This study demonstrates the contribution of gemcitabine plus nab-paclitaxel in both OS and PFS in second-line treatment of metastatic pancreatic cancer. It was found to be a good option especially for young patients with good ECOG PS.
Collapse
Affiliation(s)
- Yasin Sezgin
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey.
| | - Oğur Karhan
- Department of Medical Oncology, Faculty of Medicine, Harran University, Şanlıurfa, Turkey
| | - Mehmet Naci Aldemir
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Muslih Ürün
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Berrak Mermit Erçek
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Zuhat Urakcı
- Department of Medical Oncology, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| | - Hayati Arvas
- Department of Medical Oncology, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| | - Sezai Tunç
- Urfa Mehmet Akif Inan Regional Training Research Hospital Oncology Department, Şanlıurfa, Turkey
| | - Mehmet Erdem
- Department of Internal Medicine, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Halis Yerlikaya
- Diyarbakır private Batı Hospital Medical Oncology Department, Diyarbakır, Turkey
| | - Serdar İleri
- Gazi Yaşargil Regional Training Research Hospital Oncology Department, Diyarbakır, Turkey
| | - İbrahim Aydın
- Department of Internal Medicine, Van Regional Training Research Hospital, Van, Turkey
| | - Abdurrahman Bicer
- Department of Internal Medicine, Van Regional Training Research Hospital, Van, Turkey
| | - Ahmet Ufuk Kömüroğlu
- Health Service Vocational School of Higher Education, Van Yüzüncü Yıl University, Van, Turkey
| | - Nargiz Majidova
- Department of Medical Oncology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Savaş Gökçek
- Department of Medical Oncology, Faculty of Medicine, Eyül University, İzmir, Turkey
| | - Hacer Demir
- Department of Medical Oncology, Faculty of Medicine, Afyon Karahisar University, Afyonkarahisar, Turkey
| | - Sedat Yıldız
- Department of Medical Oncology, Faculty of Medicine, Afyon Karahisar University, Afyonkarahisar, Turkey
| | - Sinem Akbaş
- Department of Medical Oncology, Faculty of Medicine, Koç University, Istanbul, Turkey
| | - Esra Özen
- Department of Medical Oncology, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| | - Burcu Ulaş Kahya
- Department of Medical Oncology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Mürsel Sali
- Department of Medical Oncology, Faculty of Medicine, Uludağ University, Bursa, Turkey
| | - Hicran Anık
- Abdurrahman Yurtaslan Regional Training Research Hospital Oncology Department, Ankara, Turkey
| | - Talat Aykut
- Department of Medical Oncology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Murat Araz
- Department of Medical Oncology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Ali Alkan
- Department of Medical Oncology, Faculty of Medicine, Muğla Sıtkı Kocaman University, Muğla, Turkey
| | - Melike Özçelik
- Ümraniye Regional Training Research Hospital Oncology Department, Istanbul, Turkey
| | - Abudllah Sakin
- İstanbul Private Medicalpark Hospital Medical Oncology Department, Istanbul, Turkey
| | - Musa Barış Aykan
- Gülhane Regional Training Research Hospital Oncology Department, Ankara, Turkey
| | - Mirmehdi Mehtıyev
- Department of Medical Oncology, Bilkent City Hospital, Ankara, Turkey
| | - Bilgin Demir
- Faculty of Medicine, Department of Medical Oncology, Adana Menderes University, Aydın, Turkey
| | - Mehmet Nuri Başer
- Faculty of Medicine, Department of Medical Oncology, Adana Menderes University, Aydın, Turkey
| | - Müge Sönmez
- Department of Medical Oncology, Faculty of Medicine, Department of Medical Oncology, Ordu State Hospital, Sanko University, 26 İstanbul Regional, Istanbul, Turkey
| | - İlkay Gültürk
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Nilüver Avcı
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Semiha Urvay
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Mustafa Özgür Arıcı
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Mehmet Emin Kalender
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Mustafa Yıldırım
- Department of Medical Oncology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Ali Alper Solmaz
- Department of Medical Oncology, Adana City Hospital, Adana, Turkey
| | - Mustafa Gürbüz
- Department of Medical Oncology, Adana City Hospital, Adana, Turkey
| | - Yakup Ergün
- Department of Medical Oncology, Diyarbakır Private Bower Hospital, Diyarbakır, Turkey
| |
Collapse
|
2
|
Aseafan M, Alfakeeh AH, Tashkandi E, Mahrous M, Alghamdi M, Alshamsan B, Al-Hajeili M, Bakhsh S, Alshammari K, Almugbel FA, Alfagih AH, Allehebi A, Montaser M, Elsafty MH, Elnaghi KAE, Issa I, Bakshi E, AlSubaie S, AlMutairi B, Mokhtar H, Aboelatta M, Bukhari N, Alzahrani AM, Elhassan T, Alqahtani A, Bazarbashi S. Real-world clinical outcome of unresectable locally advanced & de-novo metastatic pancreatic ductal adenocarcinoma: a multicentre retrospective study. BMC Cancer 2025; 25:7. [PMID: 39754118 PMCID: PMC11697791 DOI: 10.1186/s12885-024-13386-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with limited treatment options yielding poor outcomes. This study aimed to evaluate the real-world clinical characteristics, treatment patterns, and outcomes of patients with locally advanced unresectable and de-novo metastatic PDAC in Saudi Arabia, providing regional data to compare with international benchmarks. METHODS This is a retrospective, multicentre study involving 350 patients diagnosed with unresectable locally advanced or de-novo metastatic PDAC between January 2015 and November 2023. Data were collected from 10 oncology centers across Saudi Arabia. RESULTS The median age at diagnosis was 60 years, with 63% of patients presenting with multiple metastatic sites, primarily in the liver (66.3%). FOLFIRINOX was the most common first-line treatment (55.1%), followed by gemcitabine plus nab-paclitaxel (15.1%). The median PFS for first-line treatment was 5.3 months, with FOLFIRINOX achieving the longest PFS (6.5 months). The median OS was 10.34 months for the entire cohort, with better survival outcomes observed in patients receiving FOLFIRINOX (12.3 months). Independent prognostic factors for PFS and OS included performance status, first-line regimen, and neutrophil-lymphocyte ratio (NLR). Among patients tested, 7.1% had deficient mismatch repair (d-MMR), and 5.8% harbored BRCA mutations. CONCLUSIONS This real-world study confirms that clinical outcomes for locally advanced unresectable and metastatic PDAC in Saudi Arabia are consistent with international data, with FOLFIRINOX showing superior outcomes over gemcitabine-based regimens. However, both treatments reflect the persistent poor prognosis of PDAC, underscoring the need for novel therapeutic strategies. Further research is warranted to optimize treatment selection and improve survival outcomes in this population.
Collapse
Affiliation(s)
- Mohamed Aseafan
- Section of Medical Oncology, Department of Internal Medicine, Security Forces Hospital, Riyadh, Saudi Arabia.
| | - Ali H Alfakeeh
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Emad Tashkandi
- College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mervat Mahrous
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
- Collage of Medicine, Minia University, Minia, Egypt
| | - Mohammed Alghamdi
- Oncology Center, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Bader Alshamsan
- Department of Medicine, College of Medicine, Qassim University, Buraydah, Saudi Arabia
- Prince Faisal Cancer Center, King Fahad Specialist Hospital, Qassim Health Clusster, Buraydah, Saudi Arabia
| | - Marwan Al-Hajeili
- Department of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Safwan Bakhsh
- Department of Medical Oncology, King Faisal Specialist Hospital and Research Center-Jeddah, Jeddah, Saudi Arabia
| | - Kanan Alshammari
- Department of Medical Oncology, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Fahad A Almugbel
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdulhameed H Alfagih
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ahmed Allehebi
- Department of Medical Oncology, King Faisal Specialist Hospital and Research Center-Jeddah, Jeddah, Saudi Arabia
| | - Mohamed Montaser
- Section of Medical Oncology, Department of Internal Medicine, Security Forces Hospital, Riyadh, Saudi Arabia
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Ibrahim Issa
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Eesa Bakshi
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Sadeem AlSubaie
- Pathology and Laboratory Medicine, Security Forces Hospital, Riyadh, Saudi Arabia
| | - Bandar AlMutairi
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Hoda Mokhtar
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Mohamed Aboelatta
- Prince Faisal Cancer Center, King Fahad Specialist Hospital, Qassim Health Clusster, Buraydah, Saudi Arabia
| | - Nedal Bukhari
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Ali M Alzahrani
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Tusneem Elhassan
- Research Unit, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ali Alqahtani
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Shouki Bazarbashi
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Haggstrom L, Chan WY, Nagrial A, Chantrill LA, Sim HW, Yip D, Chin V. Chemotherapy and radiotherapy for advanced pancreatic cancer. Cochrane Database Syst Rev 2024; 12:CD011044. [PMID: 39635901 PMCID: PMC11619003 DOI: 10.1002/14651858.cd011044.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is a lethal disease with few effective treatment options. Many anti-cancer therapies have been tested in the locally advanced and metastatic setting, with mixed results. This review synthesises all the randomised data available to help better inform patient and clinician decision-making. It updates the previous version of the review, published in 2018. OBJECTIVES To assess the effects of chemotherapy, radiotherapy, or both on overall survival, severe or life-threatening adverse events, and quality of life in people undergoing first-line treatment of advanced pancreatic cancer. SEARCH METHODS We searched for published and unpublished studies in CENTRAL, MEDLINE, Embase, and CANCERLIT, and handsearched various sources for additional studies. The latest search dates were in March and July 2023. SELECTION CRITERIA We included randomised controlled trials comparing chemotherapy, radiotherapy, or both with another intervention or best supportive care. Participants were required to have locally advanced, unresectable pancreatic cancer or metastatic pancreatic cancer not amenable to curative intent treatment. Histological confirmation was required. Trials were required to report overall survival. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included 75 studies in the review and 51 in the meta-analysis (11,333 participants). We divided the studies into seven categories: any anti-cancer treatment versus best supportive care; various chemotherapy types versus gemcitabine; gemcitabine-based combinations versus gemcitabine alone; various chemotherapy combinations versus gemcitabine plus nab-paclitaxel; fluoropyrimidine-based studies; miscellaneous studies; and radiotherapy studies. In general, the included studies were at low risk for random sequence generation, detection bias, attrition bias, and reporting bias, at unclear risk for allocation concealment, and high risk for performance bias. Compared to best supportive care, chemotherapy likely results in little to no difference in overall survival (OS) (hazard ratio (HR) 1.08, 95% confidence interval (CI) 0.88 to 1.33; absolute risk of death at 12 months of 971 per 1000 versus 962 per 1000; 4 studies, 298 participants; moderate-certainty evidence). The adverse effects of chemotherapy and impacts on quality of life (QoL) were uncertain. Many of the chemotherapy regimens were outdated. Eight studies compared non-gemcitabine-based chemotherapy regimens to gemcitabine. These showed that 5-fluorouracil (5FU) likely reduces OS (HR 1.69, 95% CI 1.26 to 2.27; risk of death at 12 months of 914 per 1000 versus 767 per 1000; 1 study, 126 participants; moderate certainty), and grade 3/4 adverse events (QoL not reported). Fixed dose rate gemcitabine likely improves OS (HR 0.79, 95% CI 0.66 to 0.94; risk of death at 12 months of 683 per 1000 versus 767 per 1000; 2 studies, 644 participants; moderate certainty), and likely increase grade 3/4 adverse events (QoL not reported). FOLFIRINOX improves OS (HR 0.51, 95% CI 0.43 to 0.60; risk of death at 12 months of 524 per 1000 versus 767 per 1000; P < 0.001; 2 studies, 652 participants; high certainty), and delays deterioration in QoL, but increases grade 3/4 adverse events. Twenty-eight studies compared gemcitabine-based combinations to gemcitabine. Gemcitabine plus platinum may result in little to no difference in OS (HR 0.94, 95% CI 0.81 to 1.08; risk of death at 12 months of 745 per 1000 versus 767 per 1000; 6 studies, 1140 participants; low certainty), may increase grade 3/4 adverse events, and likely worsens QoL. Gemcitabine plus fluoropyrimidine improves OS (HR 0.88, 95% CI 0.81 to 0.95; risk of death at 12 months of 722 per 1000 versus 767 per 1000; 10 studies, 2718 participants; high certainty), likely increases grade 3/4 adverse events, and likely improves QoL. Gemcitabine plus topoisomerase inhibitors result in little to no difference in OS (HR 1.01, 95% CI 0.87 to 1.16; risk of death at 12 months of 770 per 1000 versus 767 per 1000; 3 studies, 839 participants; high certainty), likely increases grade 3/4 adverse events, and likely does not alter QoL. Gemcitabine plus taxane result in a large improvement in OS (HR 0.71, 95% CI 0.62 to 0.81; risk of death at 12 months of 644 per 1000 versus 767 per 1000; 2 studies, 986 participants; high certainty), and likely increases grade 3/4 adverse events and improves QoL. Nine studies compared chemotherapy combinations to gemcitabine plus nab-paclitaxel. Fluoropyrimidine-based combination regimens improve OS (HR 0.79, 95% CI 0.70 to 0.89; risk of death at 12 months of 542 per 1000 versus 628 per 1000; 6 studies, 1285 participants; high certainty). The treatment arms had distinct toxicity profiles, and there was little to no difference in QoL. Alternative schedules of gemcitabine plus nab-paclitaxel likely result in little to no difference in OS (HR 1.10, 95% CI 0.82 to 1.47; risk of death at 12 months of 663 per 1000 versus 628 per 1000; 2 studies, 367 participants; moderate certainty) or QoL, but may increase grade 3/4 adverse events. Four studies compared fluoropyrimidine-based combinations to fluoropyrimidines alone, with poor quality evidence. Fluoropyrimidine-based combinations are likely to result in little to no impact on OS (HR 0.84, 95% CI 0.61 to 1.15; risk of death at 12 months of 765 per 1000 versus 704 per 1000; P = 0.27; 4 studies, 491 participants; moderate certainty) versus fluoropyrimidines alone. The evidence suggests that there was little to no difference in grade 3/4 adverse events or QoL between the two groups. We included only one radiotherapy (iodine-125 brachytherapy) study with 165 participants. The evidence is very uncertain about the effect of radiotherapy on outcomes. AUTHORS' CONCLUSIONS Combination chemotherapy remains standard of care for metastatic pancreatic cancer. Both FOLFIRINOX and gemcitabine plus a taxane improve OS compared to gemcitabine alone. Furthermore, the evidence suggests that fluoropyrimidine-based combination chemotherapy regimens improve OS compared to gemcitabine plus nab-paclitaxel. The effects of radiotherapy were uncertain as only one low-quality trial was included. Selection of the most appropriate chemotherapy for individuals still remains unpersonalised, with clinicopathological stratification remaining elusive. Biomarker development is essential to assist in rationalising treatment selection for patients.
Collapse
Affiliation(s)
- Lucy Haggstrom
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Wei Yen Chan
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Sydney, Australia
| | - Adnan Nagrial
- The Crown Princess Mary Cancer Centre, Westmead, Australia
- Medical School, The University of Sydney, Sydney, Australia
| | - Lorraine A Chantrill
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- University of Wollongong, Wollongong, Australia
| | - Hao-Wen Sim
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Desmond Yip
- Department of Medical Oncology, The Canberra Hospital, Garran, Australia
- ANU Medical School, Australian National University, Acton, Australia
| | - Venessa Chin
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- Medical Oncology, Garvan Institute of Medical Research, Sydney, Australia
| |
Collapse
|
4
|
Wang Q, Tong F, Qiao L, Qi L, Sun Y, Zhu Y, Ni J, Liu J, Kong W, Liu B, Du J. Hypofractionated radiotherapy plus PD-1 antibody and SOX chemotherapy as second-line therapy in metastatic pancreatic cancer: a single-arm, phase II clinical trial. Cancer Immunol Immunother 2024; 73:201. [PMID: 39105880 PMCID: PMC11303639 DOI: 10.1007/s00262-024-03744-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/21/2024] [Indexed: 08/07/2024]
Abstract
PURPOSE To assess the efficacy and safety of concurrent hypofractionated radiotherapy plus anti-PD-1 antibody and SOX chemotherapy in the treatment of metastatic pancreatic cancer (mPC) after failure of first-line chemotherapy. METHODS Patients with pathologically confirmed mPC who failed standard first-line chemotherapy were enrolled. The patients were treated with a regimen of hypofractionated radiotherapy, SOX chemotherapy, and immune checkpoint inhibitors at our institution. We collected the patients' clinical information and outcome measurements. The median progression-free survival (mPFS) was the primary endpoint of the study, followed by disease control rate (DCR), objective response rate (ORR), median overall survival (mOS) and safety. Exploratory analyses included biomarkers related to the benefits. RESULTS Between February 24, 2021, and August 30, 2023, twenty-five patients were enrolled in the study, and twenty-three patients who received at least one dose of the study agent had objective efficacy evaluation. The mPFS was 5.48 months, the mOS was 6.57 months, and the DCR and ORR were 69.5% and 30.4%, respectively. Among the seven patients who achieved a PR, the median duration of the response was 7.41 months. On-treatment decreased serum CA19-9 levels were associated with better overall survival. Besides, pretreatment inflammatory markers were associated with tumor response and survival. CONCLUSIONS Clinically meaningful antitumor activity and favorable safety profiles were demonstrated after treatment with these combination therapies in patients with refractory mPC. On-treatment decreased serum CA19-9 levels and pretreatment inflammatory markers platelet-to-lymphocyte ratio (PLR), lymphocyte-to-monocyte ratio (LMR), lactate dehydrogenase (LDH) might be biomarkers related to clinical benefits. CLINICAL TRIAL REGISTRATION https://www.chictr.org.cn/showproj.html?proj=130211 , identifier: ChiCTR2100049799, date of registration: 2021-08-09.
Collapse
Affiliation(s)
- Qin Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Fan Tong
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Li Qiao
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Liang Qi
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Yi Sun
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Yahui Zhu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Jiayao Ni
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Juan Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Weiwei Kong
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China.
- Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing, University&Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China.
| | - Juan Du
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China.
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, 210008, China.
- Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing, University&Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
5
|
Modest DP, Heinemann V, Schütt P, Angermeier S, Haberkorn M, Waidmann O, Graeven U, Wille K, Kunzmann V, Henze L, Constantin C, de Wit M, Denzlinger C, Ballhausen A, Kurreck A, Jelas I, Alig AHS, Stahler A, Stintzing S, Oettle H. Sequential therapy of refractory metastatic pancreatic cancer with 5-FU/LV/irinotecan (FOLFIRI) vs. 5-FU/LV/oxaliplatin (OFF). The PANTHEON trial (AIO PAK 0116). J Cancer Res Clin Oncol 2024; 150:332. [PMID: 38951245 PMCID: PMC11217046 DOI: 10.1007/s00432-024-05827-x] [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: 04/02/2024] [Accepted: 05/31/2024] [Indexed: 07/03/2024]
Abstract
PURPOSE In patients with metastatic pancreatic cancer, after failure of gemcitabine/nab-paclitaxel, this trial compares the efficacy of second-line therapy with FOLFIRI vs. OFF (1:1 randomisation) with cross-over to the vice-versa regimen as third-line therapy. PATIENTS AND METHODS The primary endpoint was PFS (progression-free survival: time from randomization until progression or death) of second-line therapy. The trial aimed to demonstrate non-inferiority of FOLFIRI vs OFF (non-inferiority margin of a hazard ratio (HR) of 1.5, power of 80% and a significance level of 5%, 196 events needed). Secondary endpoints included overall survival (OS), progression-free survival of third-line therapy and safety. The trial is registered with EudraCT Nr. 2016-004640-11. RESULTS The trial was terminated with 60 evaluable (37 with FOLFIRI, 23 with OFF) patients due to insufficient recruitment. PFS of second-line therapy was 2.4 (95% CI 2.3-2.6) months with FOLFIRI vs 2.4 (95% CI 2.2-2.7) months with OFF (HR: 0.80, 95% CI 0.45-1.42, P = 0.43). OS was comparable between the arms (HR: 0.95, 95% CI 0.54-1.66), P = 0.84). Only 4 out of 28 (14%) patients receiving third-line therapy achieved a disease control (partial remission or stable disease). Both second-line regimens were well tolerated without new or unexpected safety signals being observed. CONCLUSION The exploratory analysis of this early terminated trial suggests that FOLFIRI and OFF have similar efficacy ant toxicity as second-line therapy of PDAC after failure of gemcitabine/nab-paclitaxel. Third-line therapy regardless of regimen does not provide satisfactory efficacy in this sequential treatment algorithm.
Collapse
Affiliation(s)
- Dominik Paul Modest
- Charité - Universitätsmedizin Berlin, Department of Hematology, Oncology, and Cancer Immunology, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany.
| | - Volker Heinemann
- German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany
- Department of Medicine III & Comprehensive Cancer Center, Hospital of the University (LMU), Munich, Germany
| | | | - Stefan Angermeier
- Department of Gastroenterology, Hematology and Oncology, Hospital Ludwigsburg, Ludwigsburg, Germany
| | | | - Oliver Waidmann
- Centrum für Hämatologie und Onkologie Bethanien, Im Prüfling 17-19, 60389, Frankfurt, Germany
| | - Ullrich Graeven
- Department of Hematology, Oncology and Gastroenterology, Klinken Maria Hilf GmbH, Mönchengladbach, Germany
| | - Kai Wille
- University Clinic for Haematology, Oncology, Haemostaseology and Palliative Care, Johannes Wesling Medical Center Minden, University of Bochum, Bochum, Germany
| | - Volker Kunzmann
- Medical Clinic and Polyclinic II - IOT, University Hospital Würzburg, Würzburg, Germany
| | - Larissa Henze
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Christian Constantin
- Department for Oncology and Hematology, Clinical Center Lippe-Lemgo, Lemgo, Germany
| | - Maike de Wit
- Department for Internal Medicine-Hematology and Oncology, VIVANTES Hospital Neukölln, Berlin, Germany
| | | | - Alexej Ballhausen
- Charité - Universitätsmedizin Berlin, Department of Hematology, Oncology, and Cancer Immunology, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Annika Kurreck
- Charité - Universitätsmedizin Berlin, Department of Hematology, Oncology, and Cancer Immunology, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Ivan Jelas
- Charité - Universitätsmedizin Berlin, Department of Hematology, Oncology, and Cancer Immunology, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Annabel Helga Sophie Alig
- Charité - Universitätsmedizin Berlin, Department of Hematology, Oncology, and Cancer Immunology, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Arndt Stahler
- Charité - Universitätsmedizin Berlin, Department of Hematology, Oncology, and Cancer Immunology, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Sebastian Stintzing
- Charité - Universitätsmedizin Berlin, Department of Hematology, Oncology, and Cancer Immunology, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Helmut Oettle
- Practice for Internal Medicine, Joint Practice and Day Clinic, Friedrichshafen, Germany
| |
Collapse
|
6
|
Long SA, Amparo AM, Goodhart G, Ahmad SA, Waters AM. Evaluation of KRAS inhibitor-directed therapies for pancreatic cancer treatment. Front Oncol 2024; 14:1402128. [PMID: 38800401 PMCID: PMC11116577 DOI: 10.3389/fonc.2024.1402128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Despite significant advancements in the treatment of other cancers, pancreatic ductal adenocarcinoma (PDAC) remains one of the world's deadliest cancers. More than 90% of PDAC patients harbor a Kirsten rat sarcoma (KRAS) gene mutation. Although the clinical potential of anti-KRAS therapies has long been realized, all initial efforts to target KRAS were unsuccessful. However, with the recent development of a new generation of KRAS-targeting drugs, multiple KRAS-targeted treatment options for patients with PDAC have entered clinical trials. In this review, we provide an overview of current standard of care treatment, describe RAS signaling and the relevance of KRAS mutations, and discuss RAS isoform- and mutation-specific differences. We also evaluate the clinical efficacy and safety of mutation-selective and multi-selective inhibitors, in the context of PDAC. We then provide a comparison of clinically relevant KRAS inhibitors to second-line PDAC treatment options. Finally, we discuss putative resistance mechanisms that may limit the clinical effectiveness of KRAS-targeted therapies and provide a brief overview of promising therapeutic approaches in development that are focused on mitigating these resistance mechanisms.
Collapse
Affiliation(s)
- Szu-Aun Long
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Amber M. Amparo
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Grace Goodhart
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Syed A. Ahmad
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Andrew M. Waters
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
7
|
Enzler T, Nguyen A, Misleh J, Cline VJ, Johns M, Shumway N, Paulson S, Siegel R, Larson T, Messersmith W, Richards D, Chaves J, Pierce E, Zalupski M, Sahai V, Orr D, Ruste SA, Haun A, Kawabe T. A multicenter, randomized phase 2 study to establish combinations of CBP501, cisplatin and nivolumab for ≥3rd-line treatment of patients with advanced pancreatic adenocarcinoma. Eur J Cancer 2024; 201:113950. [PMID: 38422585 DOI: 10.1016/j.ejca.2024.113950] [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: 02/04/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND There is no standard of care for ≥ 3rd-line treatment of metastatic pancreatic adenocarcinoma (PDAC). CBP501 is a novel calmodulin-binding peptide that has been shown to enhance the influx of platinum agents into tumor cells and tumor immunogenicity. This study aimed to (1) confirm efficacy of CBP501/cisplatin/nivolumab for metastatic PDAC observed in a previous phase 1 study, (2) identify combinations that yield 35% 3-month progression-free survival rate (3MPFS) and (3) define the contribution of CBP501 to the effects of combination therapy. METHODS CBP501 16 or 25 mg/m2 (CBP(16) or CBP(25)) was combined with 60 mg/m2 cisplatin (CDDP) and 240 mg nivolumab (nivo), administered at 3-week intervals. Patients were randomized 1:1:1:1 to (1) CBP(25)/CDDP/nivo, (2) CBP(16)/CDDP/nivo, (3) CBP(25)/CDDP and (4) CDDP/nivo, with randomization stratified by ECOG PS and liver metastases. A Fleming two-stage design was used, yielding a one-sided type I error rate of 2.5% and 80% power when the true 3MPFS is 35%. RESULTS Among 36 patients, 3MPFS was 44.4% in arms 1 and 2, 11.1% in arm 3% and 33.3% in arm 4. Two patients achieved a partial response in arm 1 (ORR 22.2%; none in other arms). Median PFS and OS were 2.4, 2.1, 1.5 and 1.5 months and 6.3, 5.3, 3.7 and 4.9 months, respectively. Overall, all treatment combinations were well tolerated. Most treatment-related adverse events were grade 1-2. CONCLUSIONS The combination CBP(25)/(16)/CDDP/nivo demonstrated promising signs of efficacy and a manageable safety profile for the treatment of advanced PDAC. CLINICAL TRIAL REGISTRATION NCT04953962.
Collapse
Affiliation(s)
- T Enzler
- Rogel Cancer Center, University of Michigan Health, Ann Arbor, MI, USA.
| | - A Nguyen
- Comprehensive Cancer Centers of Nevada, Henderson, NV, USA
| | - J Misleh
- Medical Hematology Oncology Consultants PA, Newark, DE, USA
| | - V J Cline
- Texas Oncology - Austin Midtown, Austin, TX, USA
| | - M Johns
- Oncology Hematology Care Eastgate, Cincinnati, OH, USA
| | - N Shumway
- Texas Oncology-San Antonio Stone Oak, San Antonio, TX, USA
| | - S Paulson
- Texas Oncology - Baylor Charles A. Sammons Cancer Center, Dallas, TX, USA
| | - R Siegel
- Illinois Cancer Specialists, Arlington Heights, IL, USA
| | - T Larson
- Minnseota Oncology Hematology PA, Minneapolis, MN, USA
| | - W Messersmith
- University of Colorado Cancer Center, Aurora, CO, USA
| | - D Richards
- Texas Oncology - Northeast Texas Cancer and Research Institute, Tyler, TX, USA
| | - J Chaves
- Northwest Medical Specialties, PLLC, Tacoma, WA, USA
| | - E Pierce
- Ochsner MD Anderson Cancer Center, New Orleans, LA, USA
| | - M Zalupski
- Rogel Cancer Center, University of Michigan Health, Ann Arbor, MI, USA
| | - V Sahai
- Rogel Cancer Center, University of Michigan Health, Ann Arbor, MI, USA
| | - D Orr
- Mary Crowley Cancer Research, Dallas, TX, USA
| | - S A Ruste
- Medical Affairs, Veristat LLC, Toronto Canada
| | - A Haun
- Medical Affairs, Veristat LLC, Toronto Canada
| | - T Kawabe
- CanBas Co., Ltd., Numazu, Shizuoka, Japan
| |
Collapse
|
8
|
Denda Y, Matsuo Y, Sugita S, Eguchi Y, Nonoyama K, Murase H, Kato T, Imafuji H, Saito K, Morimoto M, Ogawa R, Takahashi H, Mitsui A, Kimura M, Takiguchi S. The Natural Product Parthenolide Inhibits Both Angiogenesis and Invasiveness and Improves Gemcitabine Resistance by Suppressing Nuclear Factor κB Activation in Pancreatic Cancer Cell Lines. Nutrients 2024; 16:705. [PMID: 38474833 PMCID: PMC10934733 DOI: 10.3390/nu16050705] [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: 01/24/2024] [Revised: 02/18/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
We previously established pancreatic cancer (PaCa) cell lines resistant to gemcitabine and found that the activity of nuclear factor κB (NF-κB) was enhanced upon the acquisition of gemcitabine resistance. Parthenolide, the main active ingredient in feverfew, has been reported to exhibit antitumor activity by suppressing the NF-κB signaling pathway in several types of cancers. However, the antitumor effect of parthenolide on gemcitabine-resistant PaCa has not been elucidated. Here, we confirmed that parthenolide significantly inhibits the proliferation of both gemcitabine-resistant and normal PaCa cells at concentrations of 10 µM and higher, and that the NF-κB activity is significantly inhibited, even by 1 µM parthenolide. In Matrigel invasion assays and angiogenesis assays, the invasive and angiogenic potentials were higher in gemcitabine-resistant than normal PaCa cells and were inhibited by a low concentration of parthenolide. Furthermore, Western blotting showed suppressed MRP1 expression in gemcitabine-resistant PaCa treated with a low parthenolide concentration. In a colony formation assay, the addition of 1 µM parthenolide improved the sensitivity of gemcitabine-resistant PaCa cell lines to gemcitabine. These results suggest that parthenolide may be used as a novel therapeutic agent for the treatment of gemcitabine-resistant PaCa.
Collapse
Affiliation(s)
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (Y.D.); (S.S.); (Y.E.); (K.N.); (H.M.); (T.K.); (H.I.); (K.S.); (M.M.); (R.O.); (H.T.); (A.M.); (M.K.); (S.T.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Wang Y, Yin Z, Gao L, Ma B, Shi J, Chen H. Lipid Nanoparticles-Based Therapy in Liver Metastasis Management: From Tumor Cell-Directed Strategy to Liver Microenvironment-Directed Strategy. Int J Nanomedicine 2023; 18:2939-2954. [PMID: 37288351 PMCID: PMC10243353 DOI: 10.2147/ijn.s402821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 05/15/2023] [Indexed: 06/09/2023] Open
Abstract
Metastasis to the liver, as one of the most frequent metastatic patterns, was associated with poor prognosis. Major drawbacks of conventional therapies in liver metastasis were the lack of metastatic-targeting ability, predominant systemic toxicities and incapability of tumor microenvironment modulations. Lipid nanoparticles-based strategies like galactosylated, lyso-thermosensitive or active-targeting chemotherapeutics liposomes have been explored in liver metastasis management. This review aimed to summarize the state-of-art lipid nanoparticles-based therapies in liver metastasis management. Clinical and translational studies on the lipid nanoparticles in treating liver metastasis were searched up to April, 2023 from online databases. This review focused not only on the updates in drug-encapsulated lipid nanoparticles directly targeting metastatic cancer cells in treating liver metastasis, but more importantly on research frontiers in drug-loading lipid nanoparticles targeting nonparenchymal liver tumor microenvironment components in treating liver metastasis, which showed promise for future clinical oncological practice.
Collapse
Affiliation(s)
- Yuhan Wang
- Lanzhou University Second Hospital, Lanzhou, 730030, People’s Republic of China
| | - Zhenyu Yin
- Lanzhou University Second Hospital, Lanzhou, 730030, People’s Republic of China
| | - Lei Gao
- Lanzhou University Second Hospital, Lanzhou, 730030, People’s Republic of China
| | - Bin Ma
- Lanzhou University Second Hospital, Lanzhou, 730030, People’s Republic of China
| | - Jianming Shi
- Lanzhou University Second Hospital, Lanzhou, 730030, People’s Republic of China
| | - Hao Chen
- Department of Surgical Oncology, Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, People’s Republic of China
| |
Collapse
|
10
|
Kondo T, Kanai M, Matsubara J, Yamaguchi D, Ura T, Kou T, Itani T, Nomura M, Funakoshi T, Yokoyama A, Doi K, Tamaoki M, Yoshimura M, Uza N, Yamada T, Masui T, Minamiguchi S, Matsumoto S, Ishikawa H, Muto M. Association between homologous recombination gene variants and efficacy of oxaliplatin-based chemotherapy in advanced pancreatic cancer: prospective multicenter observational study. Med Oncol 2023; 40:144. [PMID: 37039943 DOI: 10.1007/s12032-023-02011-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023]
Abstract
Next-generation sequencing (NGS)-based gene profiling can identify patients with pancreatic cancer with homologous recombinant repair gene pathogenic variants (HRRv). Several retrospective studies have reported a positive association between HRRv and the efficacy of platinum-based chemotherapy. However, this association remains to be validated in a prospective study. This multicenter, prospective, observational study included patients with histologically confirmed unresectable or recurrent pancreatic cancer who required systemic chemotherapy. Patients who were oxaliplatin-naïve patients were eligible. The HRRv status was measured using a College of American Pathologists-accredited NGS panel. One-year overall survival rate (1yr-OS%) was calculated after initiation of oxaliplatin-based chemotherapy and was set as the primary endpoint. Forty patients were enrolled between August 2018 and March 2020. The NGS success rate was 95% (38/40). HRRv was detected in 11 patients (27.5%). Oxaliplatin-based chemotherapy was administered to 9 of 11 patients with HRRv (81.8%) and 15 of 29 patients with non-HRRv (51.7%). The 1yr-OS% after initiation of oxaliplatin-based chemotherapy was 44.4% [95% confidence interval (CI) 13.7-71.9] and 57.1% (95% CI 28.4-78.0) in HRRv-positive and -negative cohorts, respectively. These data suggested that HRRv status alone could not be a potential predictive marker of oxaliplatin-based chemotherapy in patients with advanced pancreatic cancer. These results were in line with the results of a recent phase II study reporting the limited efficacy of poly(adenosine diphosphate-ribose) polymerase inhibitor in patients with pancreatic cancer who harbored HRRv other than BRCA. Future studies investigating patients with biallelic HRRv in the first-line setting are warranted.Trial registration UMIN000033655.
Collapse
Affiliation(s)
- Tomohiro Kondo
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Masashi Kanai
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan.
| | - Junichi Matsubara
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Daisuke Yamaguchi
- Department of Medical Oncology, Kyoto-Katsura Hospital, Kyoto, Japan
| | - Takashi Ura
- Department of Clinical Oncology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Tadayuki Kou
- Department of Gastroenterology and Hepatology, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan
| | - Toshinao Itani
- Department of Gastroenterology, Kobe City Nishi-Kobe Medical Center, Hyogo, Japan
| | - Motoo Nomura
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Taro Funakoshi
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Akira Yokoyama
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Keitaro Doi
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Masashi Tamaoki
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahiro Yamada
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
- Division of Clinical Genetics, Hokkaido University Hospital, Hokkaido, Japan
| | - Toshihiko Masui
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Shigemi Matsumoto
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Hideki Ishikawa
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Manabu Muto
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| |
Collapse
|
11
|
Irie H, Suzuki R, Okubo Y, Asama H, Konno N, Noguchi Y, Watanabe K, Shibukawa G, Imamura H, Takagi T, Sugimoto M, Sato Y, Nakamura J, Kato T, Hashimoto M, Yanagita T, Hikichi T, Ohira H. Clinical benefit of subsequent chemotherapy after drug-induced interstitial lung disease in pancreatic cancer patients: a multicenter retrospective study from Japan. BMC Cancer 2023; 23:316. [PMID: 37024781 PMCID: PMC10080831 DOI: 10.1186/s12885-023-10781-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023] Open
Abstract
PURPOSE Drug-induced interstitial lung disease (ILD) is not a rare adverse event in the current chemotherapy strategy for pancreatic ductal adenocarcinoma (PDAC). Thus, we aimed to find the optimal management for PDAC patients with a history of ILD induced by a gemcitabine-based regimen. METHODS We conducted a multicenter retrospective study. The primary endpoint was the overall survival (OS) of patients who underwent either S-1 monotherapy or FOLFOX after the onset of ILD. Toxicity data was also analyzed in the 2 groups. RESULTS Twenty-four patients were diagnosed with ILD and 17 patients who received subsequent chemotherapy were enrolled in the study. Among 17 patients who were managed with subsequent chemotherapy after recovering from ILD, we did not observe significant difference in OS between S-1 and FOLFOX (290.0 days vs. undefined, p = 0.39). Relapse of drug-induced ILD was not observed in all cases during the course. Overall, severe adverse events (CTCAE Grade 3 or 4) were observed in 3 patients (23.1%) in S-1 treatment group and 1 patient (25.0%) in FOLFOX treatment group (p = 0.93). CONCLUSIONS S-1 monotherapy and FOLFOX are comparable as the subsequent chemotherapy after gemcitabine-based chemotherapy-induced ILD in unresectable PDAC.
Collapse
Affiliation(s)
- Hiroki Irie
- Department of Gastroenterology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295 Japan
| | - Rei Suzuki
- Department of Gastroenterology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295 Japan
| | - Yoshinori Okubo
- Department of Gastroenterology, Fukushima Rosai Hospital, Fukushima, Japan
| | - Hiroyuki Asama
- Department of Gastroenterology, Fukushima Red Cross Hospital, Fukushima, Japan
| | - Naoki Konno
- Department of Gastroenterology, Fukushima Red Cross Hospital, Fukushima, Japan
| | - Yuki Noguchi
- Department of Gastroenterology, Ohta Nishinouchi Hospital, Fukushima, Japan
| | - Ko Watanabe
- Department of Gastroenterology, Ohara General Hospital, Fukushima, Japan
| | - Goro Shibukawa
- Department of Gastroenterology, Fukushima Medical University Aizu Medical Center, Fukushima, Japan
| | - Hidemichi Imamura
- Department of Gastroenterology, Ohta Nishinouchi Hospital, Fukushima, Japan
| | - Tadayuki Takagi
- Department of Gastroenterology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295 Japan
| | - Mitsuru Sugimoto
- Department of Gastroenterology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295 Japan
| | - Yuki Sato
- Department of Gastroenterology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295 Japan
| | - Jun Nakamura
- Department of Endoscopy, Fukushima Medical University Hospital, Fukushima, Japan
| | - Tsunetaka Kato
- Department of Endoscopy, Fukushima Medical University Hospital, Fukushima, Japan
| | - Minami Hashimoto
- Department of Endoscopy, Fukushima Medical University Hospital, Fukushima, Japan
| | - Takumi Yanagita
- Department of Gastroenterology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295 Japan
| | - Takuto Hikichi
- Department of Endoscopy, Fukushima Medical University Hospital, Fukushima, Japan
| | - Hiromasa Ohira
- Department of Gastroenterology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295 Japan
| |
Collapse
|
12
|
Dotan E, Catalano P, Lenchik L, Boutin R, Yao X, Marques HS, Ioffe D, Zhen DB, Li D, Wagner LI, Simon MA, Wong TZ, O'Dwyer PJ. The GIANT trial (ECOG-ACRIN EA2186) methods paper: A randomized phase II study of gemcitabine and nab-paclitaxel compared with 5-fluorouracil, leucovorin, and liposomal irinotecan in older patients with treatment-naïve metastatic pancreatic cancer - defining a new treatment option for older vulnerable patients. J Geriatr Oncol 2023; 14:101474. [PMID: 36963200 PMCID: PMC10425127 DOI: 10.1016/j.jgo.2023.101474] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/06/2023] [Indexed: 03/26/2023]
Abstract
INTRODUCTION Pancreatic cancer is the fourth leading cause of cancer-related death in the US with an increasing incidence in older adults (OA) over age 70. There are currently no treatment guidelines for OA with metastatic pancreatic cancer (mPCA) and selecting a chemotherapy regimen for these patients is subjective, based largely on chronologic age and performance status (PS). Geriatric screening tools provide a more objective and accurate evaluation of a patient's overall health but have not yet been validated in patient selection for mPCA treatment. This study aims to elucidate the optimal chemotherapy treatment of vulnerable OA with mPCA and understand the geriatric factors that affect outcomes in this population. METHODS/DESIGN The GIANT (ECOG-ACRIN EA2186) study is multicenter, randomized phase II trial enrolling patients over age 70 with newly diagnosed mPCA. This study utilizes a screening geriatric assessment (GA) which characterizes patients as fit, vulnerable, or frail. Patients with mild abnormalities in functional status and/or cognition, moderate comorbidities, or over age 80 are considered vulnerable. Enrolled patients are randomized to one of two dose-reduced treatment regimens (gemcitabine/nab-paclitaxel every other week, or dose-reduced 5-fluoruracil (5FU)/ liposomal irinotecan (nal-IRI) every other week). GA and quality of life (QoL) evaluations are completed prior to treatment initiation and at each disease evaluation. Overall survival (OS) is the primary endpoint, with secondary endpoints including progression free survival (PFS) and objective response rate (ORR). Enrolled patients will be stratified by age (70-74 vs ≥75) and ECOG PS (0-1 vs 2). Additional endpoints of interest for OA include evaluation of risk factors identified through GA, QoL evaluation, and toxicities of interest for older adults. Correlative studies include assessment of pro-inflammatory biomarkers of aging in the blood (IL-6, CRP) and imaging evaluation of sarcopenia as predictors of treatment tolerance. DISCUSSION The GIANT study is the first randomized, prospective national trial evaluating vulnerable OA with mPCA aimed at developing a tailored treatment approach for this patient population. This trial has the potential to establish a new way of objectively selecting vulnerable OA with mPCA for modified treatment and to establish a new standard of care in this growing patient population. TRIAL REGISTRATION This trial is registered with ClinicalTrial.gov Identifier NCT04233866.
Collapse
Affiliation(s)
- Efrat Dotan
- Department of Hematology and Oncology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, USA.
| | - Paul Catalano
- Dana Farber Cancer Institute - ECOG-ACRIN Biostatistics Center, Boston, MA, USA
| | - Leon Lenchik
- Department of Radiology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Robert Boutin
- Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Xin Yao
- ThedaCare Regional Cancer Center-Appelton, WI, USA
| | - Helga S Marques
- Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, RI, USA
| | - Dina Ioffe
- Department of Hematology and Oncology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, USA
| | - David B Zhen
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA; Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Daneng Li
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Lynne I Wagner
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Melissa A Simon
- Department of Obstetrics and Gynecology, Center for Health Equity Transformation, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Terence Z Wong
- Department of Radiology, Division of Nuclear Medicine and Radiotheranostics, Duke University Medical Center, Durham, NC, USA
| | - Peter J O'Dwyer
- University of Pennsylvania and Abramson Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
13
|
Fluoropyrimidine combination therapy versus fluoropyrimidine monotherapy for gemcitabine-refractory advanced pancreatic cancer: A systematic review and meta-analysis of randomized controlled trials. PLoS One 2023; 18:e0282360. [PMID: 36862702 PMCID: PMC9980826 DOI: 10.1371/journal.pone.0282360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 02/13/2023] [Indexed: 03/03/2023] Open
Abstract
OBJECTIVES Fluoropyrimidine-based regimens have been investigated as the second line chemotherapy in patients with advanced pancreatic cancer refractory to gemcitabine. We conducted this systematic review and meta-analysis to evaluate the efficacy and safety profile of fluoropyrimidine combination therapy versus fluoropyrimidine monotherapy in such patients. METHODS The databases of MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, ASCO Abstracts and ESMO Abstracts were systematically searched. Randomized controlled trials (RCTs) that compared fluoropyrimidine combination therapy versus fluoropyrimidine monotherapy in patients with gemcitabine-refractory advanced pancreatic cancer were included. The primary outcome was overall survival (OS). Secondary outcomes included progression-free survival (PFS), overall response rate (ORR) and serious toxicities. Statistical analyses were performed by using Review Manager 5.3. Egger's test was performed to assess the statistical evidence of publication bias by using stata 12.0. RESULTS A total of 1183 patients from six randomized controlled trials were included for this analysis. Fluoropyrimidine combination therapy increased ORR [RR 2.82 (1.83-4.33), p<0.00001] and PFS [HR 0.71 (0.62-0.82), p<0.00001], without significant heterogeneity. Fluoropyrimidine combination therapy improved OS [HR 0.82 (0.71-0.94), p = 0.006], with significant heterogeneity (I2 = 76%, p = 0.0009). The significant heterogeneity might have been caused by the different administration regimens and baseline characteristics. Peripheral neuropathy and diarrhea were more common in the regimens containing oxaliplatin and irinotecan, respectively. No publication bias was detected by Egger's tests. CONCLUSIONS Compared with fluoropyrimidine monotherapy, fluoropyrimidine combination therapy had a higher response rate and longer PFS in patients with gemcitabine-refractory advanced pancreatic cancer. Fluoropyrimidine combination therapy could be recommended in the second line setting. However, due to concerns about toxicities, the dose intensities of chemotherapy drugs should be carefully considered in patients with weakness.
Collapse
|
14
|
Zhang H, Tong Z, Liu L, Fu Q, Zhu X, Dai X, Bao X, Fang W, Zheng Y, Zhao P. Oxaliplatin plus irinotecan vs irinotecan as second-line treatment in pancreatic cancer patients: a randomized-controlled open-label Phase II study. Gastroenterol Rep (Oxf) 2023; 11:goac088. [PMID: 36751477 PMCID: PMC9897170 DOI: 10.1093/gastro/goac088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 11/26/2022] [Accepted: 12/08/2022] [Indexed: 02/09/2023] Open
Abstract
Background Limited second-line therapeutic options are available for metastasis pancreatic cancer (mPC). We aimed to explore the efficacy and safety of oxaliplatin plus irinotecan (IROX) in mPC patients. Methods This is an open-label, Phase 2, randomized study of mPC patients (aged 18-75 years) who failed when using gemcitabine plus S-1 as first-line therapy. Block randomization with a block size of four was used to randomly assign patients (1:1) between October 2015 and December 2017 to receive either IROX (oxaliplatin 85 mg/m2 and irinotecan 160 mg/m2) or irinotecan monotherapy (irinotecan 180 mg/m2) until disease progression, unacceptable adverse events, or consent withdrawal. The primary end point was overall survival, and the secondary end points were progression-free survival, overall response rate, and adverse event rate. Results A total of 74 patients were enrolled in this study, including 44 males and 30 females, with an average age of 61 years. The median overall survival was 10.2 and 6.7 months (adjusted hazard ratio [HR], 0.7; 95% confidence interval [CI], 0.4-1.2; P = 0.20) and the median progression-free survival was 5.1 and 2.3 months (adjusted HR, 0.4; 95% CI, 0.2-0.6; P < 0.01) in the IROX group and irinotecan group, respectively. The overall response rates were 18.4% (7/38) in the IROX group and 5.5% (2/36) in the irinotecan group (P = 0.06). Grade 3-4 adverse events occurred in 34% (13/38) of patients in the IROX group and 19% (7/36) of patients in the irinotecan group (P = 0.15). Conclusions IROX had no significant survival benefit over irinotecan monotherapy in our study. However, IROX reduced the risk of disease progression by 60%, with acceptable toxicity.
Collapse
Affiliation(s)
- Hangyu Zhang
- Department of Medical Oncology, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Zhou Tong
- Department of Medical Oncology, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Lulu Liu
- Department of Medical Oncology, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Qihan Fu
- Department of Medical Oncology, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Xudong Zhu
- Department of Medical Oncology, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Xiaomeng Dai
- Department of Medical Oncology, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Xuanwen Bao
- Department of Medical Oncology, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Weijia Fang
- Department of Medical Oncology, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Yi Zheng
- Department of Medical Oncology, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Peng Zhao
- Corresponding author. Department of Medical Oncology, School of Medicine, First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310000, P. R. China. Tel: +86-571-87235147; ;
| |
Collapse
|
15
|
Cavallazzi Sebold B, Ni G, Li J, Li H, Liu X, Wang T. PEGylated IL-10: Clinical Development in Cancer Immunotherapy, Where to Go? Curr Oncol Rep 2023; 25:115-122. [PMID: 36585961 DOI: 10.1007/s11912-022-01355-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2022] [Indexed: 01/01/2023]
Abstract
PURPOSEOF REVIEW The purpose of this review is to discuss the current understanding of the pegilodecakin (PEGylated interleukin 10) and its role in the inhibition of tumour growth and metastasis. This review also focuses on clinical data published to date that have evaluated the efficacy and safety of pegilodecakin. RECENT FINDINGS Pegilodecakin has shown significant promise in preclinical models, notable for decreased tumour burden and fewer sites of metastatic disease across various malignancies. It has been most widely assessed in a phase I/Ib clinical trial against several solid tumours, leading to the phase II and III clinical trials containing pegilodecakin and its combination with other current treatments. However, the updated data have not shown higher efficacy in renal cell carcinoma, metastatic non-small cell lung cancer or pancreatic cancer, with respect to the controls, yet the adverse events presented more mixed results. Further investigation into combination therapies including pegilodecakin is ongoing. Pegilodecakin showed promise in preclinical and phase I clinical trials on its efficacy in several solid tumours, with expected regulation of IL-10 signalling pathway observed. However, the phase II and III trials did not justify its application as potential immunotherapy in selected cancers. Further evaluation of pegilodecakin's efficacy in other cancers, either as monotherapy or in combination with the current treatments, is worth investigating clinically, which warrants to better understand its potential clinical utility.
Collapse
Affiliation(s)
- Bernardo Cavallazzi Sebold
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.,School of Science, Engineering and Technology, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia
| | - Guoying Ni
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.,The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China.,Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Junjie Li
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Hejie Li
- School of Science, Engineering and Technology, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia
| | - Xiaosong Liu
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China. .,Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China.
| | - Tianfang Wang
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia. .,School of Science, Engineering and Technology, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.
| |
Collapse
|
16
|
Strickler JH, Satake H, George TJ, Yaeger R, Hollebecque A, Garrido-Laguna I, Schuler M, Burns TF, Coveler AL, Falchook GS, Vincent M, Sunakawa Y, Dahan L, Bajor D, Rha SY, Lemech C, Juric D, Rehn M, Ngarmchamnanrith G, Jafarinasabian P, Tran Q, Hong DS. Sotorasib in KRAS p.G12C-Mutated Advanced Pancreatic Cancer. N Engl J Med 2023; 388:33-43. [PMID: 36546651 PMCID: PMC10506456 DOI: 10.1056/nejmoa2208470] [Citation(s) in RCA: 232] [Impact Index Per Article: 116.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND KRAS p.G12C mutation occurs in approximately 1 to 2% of pancreatic cancers. The safety and efficacy of sotorasib, a KRAS G12C inhibitor, in previously treated patients with KRAS p.G12C-mutated pancreatic cancer are unknown. METHODS We conducted a single-group, phase 1-2 trial to assess the safety and efficacy of sotorasib treatment in patients with KRAS p.G12C-mutated pancreatic cancer who had received at least one previous systemic therapy. The primary objective of phase 1 was to assess safety and to identify the recommended dose for phase 2. In phase 2, patients received sotorasib at a dose of 960 mg orally once daily. The primary end point for phase 2 was a centrally confirmed objective response (defined as a complete or partial response). Efficacy end points were assessed in the pooled population from both phases and included objective response, duration of response, time to objective response, disease control (defined as an objective response or stable disease), progression-free survival, and overall survival. Safety was also assessed. RESULTS The pooled population from phases 1 and 2 consisted of 38 patients, all of whom had metastatic disease at enrollment and had previously received chemotherapy. At baseline, patients had received a median of 2 lines (range, 1 to 8) of therapy previously. All 38 patients received sotorasib in the trial. A total of 8 patients had a centrally confirmed objective response (21%; 95% confidence interval [CI], 10 to 37). The median progression-free survival was 4.0 months (95% CI, 2.8 to 5.6), and the median overall survival was 6.9 months (95% CI, 5.0 to 9.1). Treatment-related adverse events of any grade were reported in 16 patients (42%); 6 patients (16%) had grade 3 adverse events. No treatment-related adverse events were fatal or led to treatment discontinuation. CONCLUSIONS Sotorasib showed anticancer activity and had an acceptable safety profile in patients with KRAS p.G12C-mutated advanced pancreatic cancer who had received previous treatment. (Funded by Amgen and others; CodeBreaK 100 ClinicalTrials.gov number, NCT03600883.).
Collapse
Affiliation(s)
- John H Strickler
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Hironaga Satake
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Thomas J George
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Rona Yaeger
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Antoine Hollebecque
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Ignacio Garrido-Laguna
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Martin Schuler
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Timothy F Burns
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Andrew L Coveler
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Gerald S Falchook
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Mark Vincent
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Yu Sunakawa
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Laetitia Dahan
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - David Bajor
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Sun-Young Rha
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Charlotte Lemech
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Dejan Juric
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Marko Rehn
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Gataree Ngarmchamnanrith
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Pegah Jafarinasabian
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Qui Tran
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - David S Hong
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| |
Collapse
|
17
|
Noel MS, Kim S, Hartley ML, Wong S, Picozzi V, Staszewski H, Kim DW, Van Tornout JM, Philip PA, Chung V, Ocean AJ, Wang‐Gillam A. A randomized phase II study of SM-88 plus methoxsalen, phenytoin, and sirolimus in patients with metastatic pancreatic cancer treated in the second line and beyond. Cancer Med 2022; 11:4169-4181. [PMID: 35499204 PMCID: PMC9678093 DOI: 10.1002/cam4.4768] [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: 12/10/2021] [Revised: 03/11/2022] [Accepted: 04/10/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This trial explores SM-88 used with methoxsalen, phenytoin, and sirolimus (MPS) in pretreated metastatic pancreatic ductal adenocarcinoma (mPDAC) METHODS: Forty-nine patients were randomized to daily 460 or 920 mg oral SM-88 with MPS (SM-88 Regimen). The primary endpoint was objective response rate (RECIST 1.1). RESULTS Thirty-seven patients completed ≥ one cycle of SM-88 Regimen (response evaluable population). Disease control rate (DCR), overall survival (OS), and progression-free survival (PFS) did not differ significantly between dose levels. Stable disease was achieved in 9/37 patients (DCR, 24.3%); there were no complete or partial responses. Quality-of-life (QOL) was maintained and trended in favor of 920 mg. SM-88 Regimen was well tolerated; a single patient (1/49) had related grade 3 and 4 adverse events, which later resolved. In the intention-to-treat population of 49 patients, the median overall survival (mOS) was 3.4 months (95% CI: 2.7-4.9 months). Those treated in the second line had an mOS of 8.1 months and a median PFS of 3.8 months. Survival was higher for patients with stable versus progressive disease (any line; mOS: 10.6 months vs. 3.9 months; p = 0.01). CONCLUSIONS SM-88 Regimen has a favorable safety profile with encouraging QOL effects, disease control, and survival trends. This regimen should be explored in the second-line treatment of patients with mPDAC. CLINICALTRIALS gov Identifier: NCT03512756.
Collapse
Affiliation(s)
- Marcus S. Noel
- Georgetown Lombardi Comprehensive Cancer CenterWashingtonDistrict of ColumbiaUSA
| | - Semmie Kim
- TYME Technologies Inc.BedminsterNew JerseyUSA
| | - Marion L. Hartley
- The Ruesch Center for the Cure of Gastrointestinal CancersWashingtonDistrict of ColumbiaUSA
| | - Steve Wong
- Sarcoma Oncology Research CenterSanta MonicaCaliforniaUSA
| | | | | | - Dae Won Kim
- The University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | | | - Philip Agop Philip
- Karmanos Cancer CenterWayne State UniversityMichiganDetroitUSA
- SWOGFarmington HillsMichiganUSA
| | | | - Allyson J. Ocean
- Weill Cornell MedicineNew York‐Presbyterian HospitalNew YorkNew YorkUSA
| | - Andrea Wang‐Gillam
- Washington University School of Medicine in St. LouisSt. LouisMissouriUSA
| |
Collapse
|
18
|
Kim G, Cockrum P, Surinach A, Wang S, Wainberg Z. Real-world safety and supportive care use of second-line 5-fluorouracil-based regimens among patients with metastatic pancreatic ductal adenocarcinoma. Curr Med Res Opin 2022; 38:1295-1303. [PMID: 35354375 DOI: 10.1080/03007995.2022.2059976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/15/2022] [Accepted: 03/28/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Chemotherapy-related adverse events (AEs) can negatively impact the care of patients. The prevention and management of AEs often require additional medications. This study evaluated the percentages of patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) undergoing second-line therapy with 5-fluorouracil (5-FU)-based regimens that experienced AEs during treatment and received medication to manage those AEs. METHODS We conducted a retrospective observational analysis utilizing the Flatiron Health database of adult patients with mPDAC who started second-line therapy between January 2016 and August 2020. The occurrence of diarrhea, fatigue, nausea and vomiting, neuropathy, and hematologic AEs including G3/G4 anemia, neutropenia, and thrombocytopenia was assessed. The use of concomitant medications including atropine and granulocyte colony stimulating factor (G-CSF) was assessed. RESULTS Of the 825 eligible patients, 29.0% (n = 239) received FOLFIRINOX, 24.0% (n = 198) received FOLFOX, 6.8% (n = 56) received FOLFIRI, and 40.2% (n = 332) received liposomal irinotecan-based regimens. FOLFIRI and FOLFIRINOX regimens were associated with the highest rates of anemia (16.1% and 15.5%), neutropenia (19.6% and 22.6%), and thrombocytopenia (14.3% and 9.6%). The liposomal irinotecan and FOLFOX regimens were associated with lower rates of anemia (11.8% and 12.1%), neutropenia (12.4% and 14.7%), and thrombocytopenia (2.4% and 8.1%). G-CSF use was observed among 63.6%, 34.9%, 33.9%, and 44.9% of patients treated with FOLFIRINOX, FOLFOX, FOLFIRI, and liposomal irinotecan-based regimens, respectively. Diarrhea was observed among 12.5%, 4.5%, 12.5%, and 10.2% of patients who were treated with FOLFIRINOX, FOLFOX, FOLFIRI, and liposomal irinotecan-based regimens, respectively. Nausea and vomiting occurred in 14.9%, 12.6%, 10.5%, and 13.1% of patients treated with FOLFIRINOX, FOLFOX, FOLFIRI, and liposomal irinotecan-based regimens, respectively. Atropine use was higher in patients treated with FOLFIRINOX and FOLFIRI (90.8% and 94.6%, respectively) than in patients treated with liposomal irinotecan-based regimens (75.6%). CONCLUSIONS In patients with mPDAC who received second-line therapy, those who received liposomal irinotecan-based regimens had the lowest rates of anemia, neutropenia, and thrombocytopenia compared to FOLFIRI, FOLFIRINOX, and FOLFOX, while requiring a similar or lower level of medication to treat and manage those adverse events. Patients treated with FOLFIRI received the highest dose of pegfilgrastim to manage neutropenia. The results of this real-world analysis are consistent with prior evaluations of patients with mPDAC and highlight the importance of managing adverse events and associated cost implications.
Collapse
Affiliation(s)
- George Kim
- Division of Hematology & Oncology, George Washington University, Washington, DC, USA
| | | | | | - Shu Wang
- Genesis Research, Hoboken, NJ, USA
| | - Zev Wainberg
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
19
|
Luo D, Liao S, Li Q, Lin Y, Wei J, Li Y, Liao X. Case Report: A Case of Locally Advanced Pancreatic Cancer Which Achieved Progression Free for Over 12 Months by Subsequent Therapy with Anlotinib Hydrochloride Plus Tegafur-Gimeracil-Oteracil Potassium (TS-1). Front Oncol 2022; 12:862600. [PMID: 35847852 PMCID: PMC9283868 DOI: 10.3389/fonc.2022.862600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/30/2022] [Indexed: 12/27/2022] Open
Abstract
Titled the "most destructive of all cancers", pancreatic cancer is a malignant tumor with a very poor prognosis and has a poor response to systemic therapy. At present, several studies have shown that tegafur-gimeracil-oteracil potassium (hereinafter referred to as TS-1) is no less superior to gemcitabine in the treatment of advanced pancreatic cancer. In addition, a number of current clinical studies have shown that targeted therapy combined with chemotherapy reflects therapeutic advantages in pancreatic cancer. Moreover, in vitro and in vivo experiments have also demonstrated that anlotinib can curb the proliferation of pancreatic cancer cells and induce their apoptosis. Here, we report for the first time that a patient with locally advanced pancreatic cancer achieved good efficacy after switching to TS-1 chemotherapy combined with anlotinib targeted therapy. Previously, the disease of the patient still rapidly progressed without control following the first switch to abraxane combined with gemcitabine chemotherapy (AG regimen) due to the progression after chemo-radiotherapy. In this case, the patient achieved progression-free survival (PFS) of over 14 months via the treatment with anlotinib targeted therapy combined with TS-1 chemotherapy and secondary radiotherapy (prior to secondary radiotherapy, the patient achieved a PFS of nearly 12 months via the treatment with oral anlotinib combined with TS-1). Up to now, the progress of the disease is ceased. The oral administration of targeted therapy and chemotherapy are still in progress and the general condition of the patient is good. This suggests that patients with advanced pancreatic cancer may benefit from treatment with the anlotinib targeted therapy combined with TS-1 chemotherapy.
Collapse
Affiliation(s)
- Dongcheng Luo
- Department of First Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Sina Liao
- Department of First Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qian Li
- Department of First Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Youzhi Lin
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Junbao Wei
- Radiotherapy Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yongqiang Li
- Department of First Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaoli Liao
- Department of First Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
20
|
Milano G, Innocenti F, Minami H. Liposomal irinotecan (Onivyde): Exemplifying the benefits of nanotherapeutic drugs. Cancer Sci 2022; 113:2224-2231. [PMID: 35445479 PMCID: PMC9277406 DOI: 10.1111/cas.15377] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 11/30/2022] Open
Abstract
Irinotecan is a topoisomerase inhibitor, widely used in treatment of malignancies including pancreatic ductal adenocarcinoma (PDAC) as part of the FOLFIRINOX regimen prescribed as a first-line treatment in several countries. However, irinotecan has not been successfully introduced as a second-line treatment for pancreatic cancer and few randomized clinical studies have evaluated its added value. Efficacy of liposomal irinotecan (nal-IRI) combined with 5-fluorouracil and leucovorin (5-FU/LV) was reported in the phase III NAPOLI-1 trial in metastatic PDAC following failure of gemcitabine-based therapy. Several features of nal-IRI pharmacokinetics (PK) could result in better outcomes versus nonliposomal irinotecan. Irinotecan is a prodrug that is converted to active SN-38 by carboxylesterase enzymes and inactivated by cytochrome P450 3A4/3A5. SN-38 is inactivated by UGT1A1 enzymes. Individual variations in their expression and activity could influence enhanced localized irinotecan activity and toxicity. Liposomal irinotecan exploits the enhanced permeability and retention effect in cancer, accumulating in tumor tissues. Liposomal irinotecan also has a longer half-life and higher area under the concentration-time curve (0-∞) than nonliposomal irinotecan, as the liposomal formulation protects cargo from premature metabolism in the plasma. This results in irinotecan activation in tumor tissue, leading to enhanced cytotoxicity. Importantly, despite the longer exposure, overall toxicity for nal-IRI is no worse than nonliposomal irinotecan. Liposomal irinotecan exemplifies how liposomal encapsulation of a chemotherapeutic agent can alter its PK properties, improving clinical outcomes for patients. Liposomal irinotecan is currently under investigation in other malignancies including biliary tract cancer (amongst other gastrointestinal cancers), brain tumors, and small-cell lung cancer.
Collapse
Affiliation(s)
- Gérard Milano
- UPR 7497Scientific Valorisation UnitCentre Antoine Lacassagne and Côte d’Azur UniversityNiceFrance
| | | | - Hironobu Minami
- Medical Oncology and HematologyKobe University Graduate School of Medicine and HospitalKobeJapan
| |
Collapse
|
21
|
Zaibet S, Hautefeuille V, Auclin E, Lièvre A, Tougeron D, Sarabi M, Gilabert M, Wasselin J, Edeline J, Artru P, Bechade D, Morin C, Ducoulombier A, Taieb J, Pernot S. Gemcitabine + Nab-paclitaxel or Gemcitabine alone after FOLFIRINOX failure in patients with metastatic pancreatic adenocarcinoma: a real-world AGEO study. Br J Cancer 2022; 126:1394-1400. [PMID: 35094032 PMCID: PMC9091233 DOI: 10.1038/s41416-022-01713-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/20/2021] [Accepted: 01/20/2022] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Gemcitabine (Gem) alone or with Nab-paclitaxel (Gem-Nab) is used as second-line treatment for metastatic pancreatic adenocarcinoma (mPA) after FOLFIRINOX (FFX) failure; however, no comparative data exist. This study evaluates the efficacy and safety of adding Nab-paclitaxel to Gem for mPA after FFX failure. METHODS In this retrospective real-world multicenter study, from 2011 to 2019, patients with mPA receiving Gem-Nab (Gem 1000 mg/m² + Nab 125 mg/m², 3 out of 4 weeks) or Gem alone were included after progression on FFX. RESULTS A total of 427 patients were included. Patients receiving Gem-Nab had more metastatic sites, peritoneal disease and less PS 2 (24% vs. 35%). After median follow-up of 22 months, Gem-Nab was associated with better disease control rate (DCR) (56% vs. 32%; P < 0.001), progression-free survival (PFS) (3.5 vs. 2.3 months; 95% CI: 0.43-0.65) and overall survival (OS) (7.1 vs. 4.7 months; 95% CI: 0.53-0.86). After multivariate analysis, Gem-Nab and PS 0/1 were associated with better OS and PFS. Grade 3/4 toxicity was more frequent with Gem-Nab (44% vs. 29%). CONCLUSION In this study, Gem-Nab was associated with better DCR, PFS and OS compared with Gem alone in patients with mPA after FFX failure, at the cost of higher toxicity.
Collapse
Affiliation(s)
- Sonia Zaibet
- Department of Hepato-Gastroenterology and Gastrointestinal Oncology, Hôpital Européen Georges Pompidou, Université de Paris, SIRIC CARPEM Comprehensive Cancer Center, Paris, France
| | - Vincent Hautefeuille
- Department of Hepato-Gastroenterology and Gastrointestinal Oncology, CHU Amiens Picardie, Amiens, France
| | - Edouard Auclin
- Department of Medical Oncology, Hôpital Européen Georges Pompidou, Université de Paris, SIRIC CARPEM Comprehensive Cancer Center, Paris, France
- INSERM, UMR 1138, team 22, Centre de Recherche des Cordeliers, Université de Paris, Paris, France
| | - Astrid Lièvre
- Department of Gastroenterology, CHU Rennes, INSERM U1242, Rennes, France
| | - David Tougeron
- Department of Hepato-Gastroenterology, CHU Poitiers, Poitiers, France
| | - Mathieu Sarabi
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Marine Gilabert
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Julie Wasselin
- Department of Hepato-Gastroenterology and Gastrointestinal Oncology, CHU Amiens Picardie, Amiens, France
| | - Julien Edeline
- Department of Medical Oncology, Centre Eugène Marquis, Rennes, France
| | - Pascal Artru
- Department of Hepato-Gastroenterology, Hôpital Privé Jean Mermoz, Lyon, France
| | | | - Clémence Morin
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | | | - Julien Taieb
- Department of Hepato-Gastroenterology and Gastrointestinal Oncology, Hôpital Européen Georges Pompidou, Université de Paris, SIRIC CARPEM Comprehensive Cancer Center, Paris, France
| | - Simon Pernot
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France.
| |
Collapse
|
22
|
Ueda G, Matsuo Y, Murase H, Aoyama Y, Kato T, Omi K, Hayashi Y, Imafuji H, Saito K, Tsuboi K, Morimoto M, Ogawa R, Takahashi H, Mitsui A, Kimura M, Takiguchi S. 10Z‑Hymenialdisine inhibits angiogenesis by suppressing NF‑κB activation in pancreatic cancer cell lines. Oncol Rep 2022; 47:48. [PMID: 35014682 PMCID: PMC8771196 DOI: 10.3892/or.2022.8259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022] Open
Abstract
10Z‑Hymenialdisine is a natural product derived from the marine sponge Axinella carteri. 10Z‑Hymenialdisine has anti‑inflammatory effects exerted through NF‑κB; however, it is unclear whether 10Z‑Hymenialdisine has anti‑angiogenic effects in cancer cells. In the present study, both the anti‑angiogenic and antimetastatic effects of this compound in pancreatic cancer were investigated. It was initially confirmed that 10Z‑Hymenialdisine significantly inhibited the proliferation of pancreatic cancer cells. Next, using both reverse transcription‑quantitative PCR and ELISA, it was demonstrated that 10Z‑Hymenialdisine significantly suppressed the expression of VEGF and IL‑8 mRNAs and proteins in pancreatic cancer. Immunohistochemical analysis revealed that 10Z‑Hymenialdisine inhibited NF‑κB activity in pancreatic cancer cell lines. It was also identified that 10Z‑Hymenialdisine inhibited tube formation in EA.hy926 cells. In vivo, 10Z‑Hymenialdisine significantly inhibited the growth of BxPC‑3 pancreatic cancer cells that were subcutaneously injected into model mice. In conclusion, the present study demonstrated that 10Z‑Hymenialdisine exerted anti‑angiogenic effects by suppressing NF‑κB activity and angiogenic factors, such as VEGF and IL‑8, in pancreatic cancer cell lines. 10Z‑Hymenialdisine has potential applications as a novel therapeutic agent for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Goro Ueda
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Hiromichi Murase
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Yoshinaga Aoyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Tomokatsu Kato
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Kan Omi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Yuichi Hayashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Hiroyuki Imafuji
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Kenta Saito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Ken Tsuboi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
- Department of Gastrointestinal Surgery, Nagoya City East Medical Center, Chikusa-ku, Nagoya, Aichi 464-8547, Japan
| | - Mamoru Morimoto
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Akira Mitsui
- Department of Gastroenterological Surgery, Nagoya City University West Medical Center, Kita-ku, Nagoya, Aichi 462-8508, Japan
| | - Masahiro Kimura
- Department of Gastrointestinal Surgery, Nagoya City East Medical Center, Chikusa-ku, Nagoya, Aichi 464-8547, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences and Medical School, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
23
|
Huh G, Lee HS, Choi JH, Lee SH, Paik WH, Ryu JK, Kim YT, Bang S, Lee ES. Gemcitabine plus Nab-paclitaxel as a second-line treatment following FOLFIRINOX failure in advanced pancreatic cancer: a multicenter, single-arm, open-label, phase 2 trial. Ther Adv Med Oncol 2021; 13:17588359211056179. [PMID: 34790261 PMCID: PMC8591648 DOI: 10.1177/17588359211056179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/11/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The aim of this study was to evaluate the efficacy and safety of gemcitabine plus nab-paclitaxel (GnP) as second-line chemotherapy following first-line FOLFIRINOX treatment failure in advanced pancreatic cancer. METHODS This was a multicenter, single-arm, open-label, phase 2 trial done at three tertiary centers in South Korea from May 2018 to December 2019. Eligible patients were aged 20 years or older, had histologically confirmed advanced pancreatic ductal adenocarcinoma, and disease progression after receiving first-line FOLFIRINOX. Patients received a second-line GnP regimen as intravenous nab-paclitaxel at a dose of 125 mg/m2 and gemcitabine at a dose of 1000 mg/m2, on days 1, 8, and 15 every 4 weeks until disease progression or unacceptable toxicity. The primary outcome was survival rate at 6 months and the secondary outcomes were median progression-free survival (PFS), overall survival (OS), disease control rate (DCR), and adverse events. This study is registered with Clinicaltrials.gov. (NCT03401827). RESULTS Forty patients were enrolled in the study. The survival rate at 6 months was 72.5% [95% confidence interval (CI), 59.9-87.7], achieving superiority over prespecified assumed 6-month OS rate of 20% for best supportive care only (p < 0.001). The median PFS and OS were 5.8 months (95% CI, 4.3-8.7) and 9.9 months (95% CI, 7.5-12.4), respectively. DCR was 87.5% with six partial responses and 29 stable diseases. Grade 3 or higher treatment-related adverse events occurred in 25 (62.5%) patients with the most common being thrombocytopenia, anemia, neutropenia, peripheral neuropathy, and peripheral edema. CONCLUSION GnP demonstrated favorable efficacy with acceptable toxicity in patients with advanced pancreatic ductal adenocarcinoma after FOLFIRINOX failure.
Collapse
Affiliation(s)
- Gunn Huh
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
- Pancreaticobiliary Cancer Study Group of Korean Society of Gastrointestinal Cancer, Seoul, Korea
| | - Hee Seung Lee
- Pancreaticobiliary Cancer Study Group of Korean Society of Gastrointestinal Cancer, Seoul, Korea
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Ho Choi
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
- Pancreaticobiliary Cancer Study Group of Korean Society of Gastrointestinal Cancer, Seoul, Korea
| | - Sang Hyub Lee
- Professor, Division of Gastroenterology, Department of Internal Medicine, Seoul National University College of Medicine, Liver Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
- Pancreaticobiliary Cancer Study Group of Korean Society of Gastrointestinal Cancer, Seoul, Korea
| | - Woo Hyun Paik
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
- Pancreaticobiliary Cancer Study Group of Korean Society of Gastrointestinal Cancer, Seoul, Korea
| | - Ji Kon Ryu
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
- Pancreaticobiliary Cancer Study Group of Korean Society of Gastrointestinal Cancer, Seoul, Korea
| | - Yong-Tae Kim
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
- Pancreaticobiliary Cancer Study Group of Korean Society of Gastrointestinal Cancer, Seoul, Korea
| | - Seungmin Bang
- Pancreaticobiliary Cancer Study Group of Korean Society of Gastrointestinal Cancer, Seoul, Korea
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Eaum Seok Lee
- Pancreaticobiliary Cancer Study Group of Korean Society of Gastrointestinal Cancer, Seoul, Korea
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
24
|
Stoof J, Harrold E, Mariottino S, Lowery MA, Walsh N. DNA Damage Repair Deficiency in Pancreatic Ductal Adenocarcinoma: Preclinical Models and Clinical Perspectives. Front Cell Dev Biol 2021; 9:749490. [PMID: 34712667 PMCID: PMC8546202 DOI: 10.3389/fcell.2021.749490] [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: 07/29/2021] [Accepted: 09/22/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide, and survival rates have barely improved in decades. In the era of precision medicine, treatment strategies tailored to disease mutations have revolutionized cancer therapy. Next generation sequencing has found that up to a third of all PDAC tumors contain deleterious mutations in DNA damage repair (DDR) genes, highlighting the importance of these genes in PDAC. The mechanisms by which DDR gene mutations promote tumorigenesis, therapeutic response, and subsequent resistance are still not fully understood. Therefore, an opportunity exists to elucidate these processes and to uncover relevant therapeutic drug combinations and strategies to target DDR deficiency in PDAC. However, a constraint to preclinical research is due to limitations in appropriate laboratory experimental models. Models that effectively recapitulate their original cancer tend to provide high levels of predictivity and effective translation of preclinical findings to the clinic. In this review, we outline the occurrence and role of DDR deficiency in PDAC and provide an overview of clinical trials that target these pathways and the preclinical models such as 2D cell lines, 3D organoids and mouse models [genetically engineered mouse model (GEMM), and patient-derived xenograft (PDX)] used in PDAC DDR deficiency research.
Collapse
Affiliation(s)
- Jojanneke Stoof
- Trinity St. James Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Emily Harrold
- Trinity College Dublin, Dublin, Ireland
- Mater Private Hospital, Dublin, Ireland
| | - Sarah Mariottino
- Trinity St. James Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Maeve A Lowery
- Trinity St. James Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Naomi Walsh
- National Institute of Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
25
|
Kamgar M, Chakrabarti S, Shreenivas A, George B. Evolution of Systemic Therapy in Metastatic Pancreatic Ductal Adenocarcinoma. Surg Oncol Clin N Am 2021; 30:673-691. [PMID: 34511189 DOI: 10.1016/j.soc.2021.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Pancreatic ductal adenocarcinoma is characterized by early systemic dissemination, a complex tumor microenvironment, as well as significant intratumoral and intertumoral heterogeneity. Treatment options and survival in pancreatic ductal adenocarcinoma have improved steadily over the last 3 decades. Although cytotoxic chemotherapy is currently the mainstay of treatment for pancreatic ductal adenocarcinoma, evolving therapeutic strategies are aimed at targeting the tumor microenvironment, metabolism, and the tumor-host immune balance.
Collapse
Affiliation(s)
- Mandana Kamgar
- Division of Hematology and Oncology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA.
| | - Sakti Chakrabarti
- Division of Hematology and Oncology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA
| | - Aditya Shreenivas
- Division of Hematology and Oncology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA
| | - Ben George
- Division of Hematology and Oncology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA
| |
Collapse
|
26
|
Winer A, Dotan E. Treatment Paradigms for Older Adults with Pancreatic Cancer: a Nuanced Approach. Curr Treat Options Oncol 2021; 22:104. [PMID: 34596801 DOI: 10.1007/s11864-021-00892-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer is increasing in incidence in the USA. This disease disproportionately affects older adults, and as the percentage of adults > 65 years old increases with the aging of the baby boomers, the prevalence is expected to rise over the coming decade. These patients are often more susceptible to disease-related symptoms and have less ability to withstand both cancer and treatment-related side effects. Therefore, it is imperative that treating physicians thoughtfully consider their recommended treatment approach towards this vulnerable patient population. This review focuses on the current state of research of older adults with pancreatic adenocarcinoma, highlighting deficiencies in the representation of this patient population in clinical trials. It is vital that the treating physicians take a nuanced approach towards therapy of localized and metastatic disease in geriatric patients. A one size fits all treatment algorithm is no longer appropriate in any cancer patient, let alone the elders who are particularly vulnerable to developing treatment-related toxicities. To help guide therapy decisions, it is important to perform a comprehensive geriatric assessment which may uncover unexpected frailty and lead to a change in the recommended treatment approach. In this way, we can support older adults during therapy for this aggressive malignancy and provide optimal care.
Collapse
Affiliation(s)
- Arthur Winer
- Department of Medical Oncology, Inova Schar Cancer Institute, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA.
| | - Efrat Dotan
- Department of Medical Oncology, Fox Chase Cancer, Philadelphia, PA, USA
| |
Collapse
|
27
|
Pijnappel EN, Wassenaar NPM, Gurney-Champion OJ, Klaassen R, van der Lee K, Pleunis-van Empel MCH, Richel DJ, Legdeur MC, Nederveen AJ, van Laarhoven HWM, Wilmink JW. Phase I/II Study of LDE225 in Combination with Gemcitabine and Nab-Paclitaxel in Patients with Metastatic Pancreatic Cancer. Cancers (Basel) 2021; 13:4869. [PMID: 34638351 PMCID: PMC8507646 DOI: 10.3390/cancers13194869] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Desmoplasia is a central feature of the tumor microenvironment in pancreatic ductal adenocarcinoma (PDAC). LDE225 is a pharmacological Hedgehog signaling pathway inhibitor and is thought to specifically target tumor stroma. We investigated the combined use of LDE225 and chemotherapy to treat PDAC patients. METHODS This was a multi-center, phase I/II study for patients with metastatic PDAC establishing the maximum tolerated dose of LDE225 co-administered with gemcitabine and nab-paclitaxel (phase I) and evaluating the efficacy and safety of the treatment combination after prior FOLFIRINOX treatment (phase II). Tumor microenvironment assessment was performed with quantitative MRI using intra-voxel incoherent motion diffusion weighted MRI (IVIM-DWI) and dynamic contrast-enhanced (DCE) MRI. RESULTS The MTD of LDE225 was 200 mg once daily co-administered with gemcitabine 1000 mg/m2 and nab-paclitaxel 125 mg/m2. In phase II, six therapy-related grade 4 adverse events (AE) and three grade 5 were observed. In 24 patients, the target lesion response was evaluable. Three patients had partial response (13%), 14 patients showed stable disease (58%), and 7 patients had progressive disease (29%). Median overall survival (OS) was 6 months (IQR 3.9-8.1). Blood plasma fraction (DCE) and diffusion coefficient (IVIM-DWI) significantly increased during treatment. Baseline perfusion fraction could predict OS (>222 days) with 80% sensitivity and 85% specificity. CONCLUSION LDE225 in combination with gemcitabine and nab-paclitaxel was well-tolerated in patients with metastatic PDAC and has promising efficacy after prior treatment with FOLFIRINOX. Quantitative MRI suggested that LDE225 causes increased tumor diffusion and works particularly well in patients with poor baseline tumor perfusion.
Collapse
Affiliation(s)
- Esther N. Pijnappel
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Nienke P. M. Wassenaar
- Cancer Center Amsterdam, Department of Radiology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (N.P.M.W.); (O.J.G.-C.); (A.J.N.)
| | - Oliver J. Gurney-Champion
- Cancer Center Amsterdam, Department of Radiology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (N.P.M.W.); (O.J.G.-C.); (A.J.N.)
| | - Remy Klaassen
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Koen van der Lee
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | | | - Dick J. Richel
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Marie C. Legdeur
- Department of Medical Oncology, Medisch Spectrum Twente, Twente, 7512 Enschede, The Netherlands; (M.C.H.P.-v.E.); (M.C.L.)
| | - Aart J. Nederveen
- Cancer Center Amsterdam, Department of Radiology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (N.P.M.W.); (O.J.G.-C.); (A.J.N.)
| | - Hanneke W. M. van Laarhoven
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| | - Johanna W. Wilmink
- Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, 1012 Amsterdam, The Netherlands; (E.N.P.); (R.K.); (K.v.d.L.); (D.J.R.); (H.W.M.v.L.)
| |
Collapse
|
28
|
Wang T, He W, Du Y, Wang J, Li X. Redox-sensitive irinotecan liposomes with active ultra-high loading and enhanced intracellular drug release. Colloids Surf B Biointerfaces 2021; 206:111967. [PMID: 34256270 DOI: 10.1016/j.colsurfb.2021.111967] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/01/2021] [Accepted: 07/04/2021] [Indexed: 01/21/2023]
Abstract
In this report, a novel irinotecan (IR) encapsulated redox-responsive liposome was developed. The redox-responsive liposomes were prepared based on disulfide phosphatidylcholine (SS-PC), DSPC, DSPE-PEG2000 and cholesterol by ethanol injection method. IR was actively loaded by triethylammonium sucrose octasulfate (TEA8-SOS) gradient method to generate IR/SS-PC liposomes (IR/SS-LP). The particle size of IR/SS-PC was characterized by using dynamic light scattering (DLS) and transmission electron microscopy (TEM). It was found that IR/SS-LP with 30 % content of SS-PC (IR/SS30-LP) had an average size of 125.5 ± 5.8 nm with a negative zeta potential of -19.5 ± 0.1. The encapsulation efficiency (EE) was further determined to be 98.1 ± 0.8 % and drug loading (DL) was 31.8 ± 0.1 %. The redox-responsiveness of IR/SS-LP was investigated by observing the change of particle size and morphology as well as the release behavior of IR triggered by glutathione (GSH). The data indicated GSH breaks the disulfide bonds in SS-PC and leads to the controlled release of IR. At 1 mM GSH, 60.2 % irinotecan was released from IR/SS30-LP within 24 h. Finally, the effects of IR/SS-LP in cell and animal experiments were evaluated in detail. The results showed that IR/SS30-LP had superior pharmacokinetic and antitumor efficacy compared to free irinotecan and traditional irinotecan liposome (ONIVYDE®-like). Taken together, IR/SS30-LP displayed redox-responsive release of IR, ultra-high loading and enhanced anti-tumor activity, which has great potential for clinical application as a new generation of IR liposomal formulation.
Collapse
Affiliation(s)
- Tao Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Wei He
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Yawei Du
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Ji Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, PR China.
| |
Collapse
|
29
|
Lellouche L, Palmieri LJ, Dermine S, Brezault C, Chaussade S, Coriat R. Systemic therapy in metastatic pancreatic adenocarcinoma: current practice and perspectives. Ther Adv Med Oncol 2021; 13:17588359211018539. [PMID: 34285720 PMCID: PMC8264726 DOI: 10.1177/17588359211018539] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/29/2021] [Indexed: 12/25/2022] Open
Abstract
Major breakthroughs have been achieved in the management of metastatic pancreatic ductal adenocarcinoma (PDAC) with FOLFIRINOX (5-fluorouracil + irinotecan + oxaliplatin) and gemcitabine plus nab-paclitaxel approved as a first-line therapy, although the prognosis is still poor. At progression, patients who maintain a good performance status (PS) can benefit from second-line chemotherapy. To address the concern of achieving tumor control while maintaining a good quality of life, maintenance therapy is a concept that has now emerged. After a FOLFIRINOX induction treatment, maintenance with 5-fluorouracil (5-FU) seems to offer a promising approach. Although not confirmed in large, prospective trials, gemcitabine alone as a maintenance therapy following induction treatment with gemcitabine plus nab-paclitaxel could be an option, while a small subset of patients with a germline mutation of breast cancer gene (BRCA) can benefit from the polyadenosine diphosphate-ribose polymerase (PARP) inhibitor olaparib. The rate of PDAC with molecular alterations that could lead to a specific therapy is up to 25%. The Food and Drug Administration (FDA) recently approved larotrectinib for patients with any tumors harboring a neurotrophic tyrosine receptor kinase (NTRK) gene fusion, and pembrolizumab for patients with a mismatch repair deficiency in a second-line setting, including PDAC. Research focused on targeted therapy and immunotherapy is active and could improve patients' outcomes in the near future.
Collapse
Affiliation(s)
- Lisa Lellouche
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, APHP. Centre, Paris, France
- Faculté de Médecine Paris Centre, Université de Paris, Paris, France
| | - Lola-Jade Palmieri
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, APHP. Centre, 27 rue du faubourg St Jacques, Paris, 75014, France
- Faculté de Médecine Paris Centre, Université de Paris, Paris, 75006, France
| | - Solène Dermine
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, APHP. Centre, Paris, France
- Faculté de Médecine Paris Centre, Université de Paris, Paris, France
| | - Catherine Brezault
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, Paris, France
| | - Stanislas Chaussade
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, APHP. Centre, Paris, France
- Faculté de Médecine Paris Centre, Université de Paris, Paris, France
| | - Romain Coriat
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, APHP. Centre, Paris, France
- Faculté de Médecine Paris Centre, Université de Paris, Paris, France
| |
Collapse
|
30
|
Cherri S, Noventa S, Zaniboni A. Pancreatic adenocarcinoma: Beyond first line, where are we? World J Gastroenterol 2021; 27:1847-1863. [PMID: 34007126 PMCID: PMC8108033 DOI: 10.3748/wjg.v27.i17.1847] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/09/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is considered one of the most aggressive cancers, with an increasing incidence in recent years. To date, chemotherapy is still the standard of care for advanced metastatic disease, unfortunately providing only a slight advantage in terms of survival. The molecular and cellular characteristics of pancreatic cancer cells, as well as the cells that characterize the pancreatic tumour microenvironment, are the basis of the mechanisms of resistance to treatment. After progression during first-line treatment, few patients are eligible for second-line treatment due to the loss of performance status. To date, a clear survival advantage has not yet been demonstrated for second-line chemotherapy. Precision medicine could be the key to increasing responses to cancer treatment and finally impacting survival in this difficult-to-treat disease. In this review, we analyze current recommendations in the second-line setting and potential future prospects.
Collapse
Affiliation(s)
- Sara Cherri
- Department of Oncology, Fondazione Poliambulanza, Brescia 25124, Italy
| | - Silvia Noventa
- Department of Oncology, Fondazione Poliambulanza, Brescia 25124, Italy
| | - Alberto Zaniboni
- Department of Oncology, Fondazione Poliambulanza, Brescia 25124, Italy
| |
Collapse
|
31
|
Gutierrez-Sainz L, Viñal D, Villamayor J, Martinez-Perez D, Garcia-Cuesta JA, Ghanem I, Custodio A, Feliu J. Prognostic factors in advanced pancreatic ductal adenocarcinoma patients-receiving second-line treatment: a single institution experience. Clin Transl Oncol 2021; 23:1838-1846. [PMID: 33866520 DOI: 10.1007/s12094-021-02589-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/08/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Second-line (2L) treatments for advanced pancreatic ductal adenocarcinoma (PDAC) achieve a modest benefit at the expense of potential toxicity. In the absence of predictive factors of response, the identification of prognostic factors could help in the therapeutic decisions-making. The purpose of this study was to assess the prognostic factors associated with shorter survival in patients with advanced PDAC who received 2L treatment. METHODS We conducted a single institution retrospective study, which included all patients with advanced PDAC who received 2L treatment between September 2006 and February 2020 at La Paz University Hospital, Madrid (Spain). Significant variables in the logistic regression model were used to create a prognostic score. RESULTS We included 108 patients. The median overall survival (OS) was 5.10 months (95%CI 4.02-6.17). In the multivariate analysis, time to progression (TTP) shorter than 4 months after first-line treatment (OR 4.53 [95%CI 1.28-16.00] p = 0.01), neutrophil-to-lymphocyte ratio (NLR) greater than 3 at the beginning of 2L (OR 9.07 [95%CI 1.82-45.16] p = 0.01) and CA-19.9 level higher than the upper limit of normal at the beginning of 2L (OR 7.83 [95%CI 1.30-49.97] p = 0.02) were independently associated with OS shorter than 3 months. The prognostic score classified patients into three prognostic groups (good, intermediate and poor) with significant differences in OS (p < 0.001). CONCLUSIONS TTP shorter than 4 months after first-line treatment, NLR greater than 3 and CA-19.9 level higher than the upper limit of normal at the beginning of 2L were associated with shorter overall survival. We developed a prognostic score that classifies patients with advanced PDAC into three prognostic groups after progression to the first-line. This score could help in the decision-making for 2L treatment.
Collapse
Affiliation(s)
- L Gutierrez-Sainz
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.
| | - D Viñal
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - J Villamayor
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - D Martinez-Perez
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - J A Garcia-Cuesta
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - I Ghanem
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - A Custodio
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.,Cátedra UAM-AMGEN, Madrid, Spain.,CIBERONC, Madrid, Spain
| | - J Feliu
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.,Cátedra UAM-AMGEN, Madrid, Spain.,CIBERONC, Madrid, Spain
| |
Collapse
|
32
|
Hecht JR, Lonardi S, Bendell J, Sim HW, Macarulla T, Lopez CD, Van Cutsem E, Muñoz Martin AJ, Park JO, Greil R, Wang H, Hozak RR, Gueorguieva I, Lin Y, Rao S, Ryoo BY. Randomized Phase III Study of FOLFOX Alone or With Pegilodecakin as Second-Line Therapy in Patients With Metastatic Pancreatic Cancer That Progressed After Gemcitabine (SEQUOIA). J Clin Oncol 2021; 39:1108-1118. [PMID: 33555926 PMCID: PMC8078437 DOI: 10.1200/jco.20.02232] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/26/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
PURPOSE SEQUOIA compared efficacy and safety of adding pegilodecakin (PEG), a pegylated recombinant human interleukin (IL)-10, with folinic acid, fluorouracil, and oxaliplatin (FOLFOX) in patients following progression on first-line gemcitabine-containing therapy with metastatic pancreatic ductal adenocarcinoma (PDAC). PATIENTS AND METHODS SEQUOIA, a randomized, global phase III study, compared FOLFOX with PEG + FOLFOX as second line in gemcitabine-refractory PDAC. Patients were randomly assigned 1:1 (PEG + FOLFOX:FOLFOX) and stratified by prior gemcitabine and region. Eligible patients had only one prior gemcitabine-containing treatment. Primary end point was overall survival (OS). Secondary end points included progression-free survival (PFS), response evaluation per Response Evaluation Criteria in Solid Tumor (RECIST) 1.1, and safety. Exploratory analyses included biomarkers related to immune activation. RESULTS Between March 1, 2017, and September 9, 2019, 567 patients were randomly assigned PEG + FOLFOX (n = 283) or FOLFOX (n = 284). Most (94.7%) patients received prior gemcitabine plus nab paclitaxel. OS was similar comparing PEG + FOLFOX versus FOLFOX (median: 5.8 v 6.3 months; hazard ratio = 1.045; 95% CI, 0.863 to 1.265). Also, PFS (median 2.1 v 2.1 months; hazard ratio = 0.981; 95% CI, 0.808 to 1.190) and objective response rate (4.6% v 5.6%) were similar between the treatment arms. Most common (≥ 35%) treatment-emergent adverse events in PEG + FOLFOX versus FOLFOX were thrombocytopenia (55% v 20%), anemia (40% v 16%), fatigue (61% v 45%), neutropenia (39% v 28%), abdominal pain (37% v 29%), nausea (45% v 41%), neuropathy (37% v 38%), and decreased appetite (35% v 31%). Exploratory analyses revealed increases in total IL-18, interferon (IFN)-γ, and granzyme B and decreases in transforming growth factor (TGF)-β with the addition of PEG. CONCLUSION PEG added to FOLFOX did not improve efficacy in advanced gemcitabine-refractory PDAC. Safety findings were consistent as previously observed from PEG with chemotherapy; toxicity was manageable and tolerable. Exploratory pharmacodynamic results were consistent with immunostimulatory signals of the IL-10R pathway.
Collapse
Affiliation(s)
| | | | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Hao-Wen Sim
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Teresa Macarulla
- Vall dʹHebrón University Hospital and Vall dʹHebrón Institute of Oncology (VHIO), IOB Quirón, Barcelona, Spain
| | | | | | - Andres J. Muñoz Martin
- Instituto de Investigaciόn Sanitaria Gregorio Marañόn, Universidad Complutense, Madrid, Spain
| | - Joon Oh Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Richard Greil
- Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute-CCCIT and Cancer Cluster Salzburg, Salzburg, Austria
| | - Hong Wang
- Eli Lilly and Company, Indianapolis, IN
| | | | | | - Yong Lin
- Eli Lilly and Company, New York, NY
| | | | - Baek-Yeol Ryoo
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
33
|
Park HS, Kang B, Chon HJ, Im HS, Lee CK, Kim I, Kang MJ, Hwang JE, Bae WK, Cheon J, Park JO, Hong JY, Kang JH, Kim JH, Lim SH, Kim JW, Kim JW, Yoo C, Choi HJ. Liposomal irinotecan plus fluorouracil/leucovorin versus FOLFIRINOX as the second-line chemotherapy for patients with metastatic pancreatic cancer: a multicenter retrospective study of the Korean Cancer Study Group (KCSG). ESMO Open 2021; 6:100049. [PMID: 33578192 PMCID: PMC7878976 DOI: 10.1016/j.esmoop.2021.100049] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/31/2020] [Accepted: 01/03/2021] [Indexed: 12/16/2022] Open
Abstract
Background There is no clear consensus on the recommended second-line treatment for patients with metastatic pancreatic cancer who have disease progression following gemcitabine-based therapy. We retrospectively evaluated the clinical outcomes of liposomal irinotecan (nal-IRI) plus fluorouracil/leucovorin (FL) and FOLFIRINOX (fluorouracil, leucovorin, irinotecan, and oxaliplatin) in patients who had failed on the first-line gemcitabine-based therapy. Patients and methods From January 2015 to August 2019, 378 patients with MPC who had received nal-IRI/FL (n = 104) or FOLFIRINOX (n = 274) as second-line treatment across 11 institutions were included in this retrospective study. Results There were no significant differences in baseline characteristics between groups, except age and first-line regimens. With a median follow-up of 6 months, the median progression-free survival (PFS) was 3.7 months with nal-IRI/FL versus 4.6 months with FOLFIRINOX (P = 0.44). Median overall survival (OS) was 7.7 months with nal-IRI/FL versus 9.7 months with FOLFRINOX (P = 0.13). There was no significant difference in PFS and OS between the two regimens in the univariate and multivariate analyses. The subgroup analysis revealed that younger age (<70 years) was associated with better OS with FOLFIRINOX. In contrast, older age (≥70 years) was associated with better survival outcomes with nal-IRI/FL. Adverse events were manageable with both regimens; however, the incidence of grade 3 or higher neutropenia and peripheral neuropathy was higher in patients treated with FOLFIRINOX than with nal-IRI/FL. Conclusions Second-line nal-IRI/FL and FOLFIRINOX showed similar effectiveness outcomes after progression following first-line gemcitabine-based therapy. Age could be the determining factor for choosing the appropriate second-line therapy. This multicenter retrospective study investigated nal-IRI/FL and FOLFIRINOX outcomes after gemcitabine-based therapy. We found no significant differences in outcome between nal-IRI/FL and FOLFIRINOX treatment. Both regimens were well tolerated; however, neutropenia and peripheral neuropathy were more frequent with FOLFIRINOX. Age (cut-off, 70 years) showed differential efficacy between chemotherapy regimens.
Collapse
Affiliation(s)
- H S Park
- Division of Medical Oncology, Department of Internal Medicine, St. Vincent's Hospital, The Catholic University of Korea, Seoul, Korea
| | - B Kang
- Medical Oncology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - H J Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - H-S Im
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - C-K Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - I Kim
- Department of Internal Medicine, Inje University Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - M J Kang
- Department of Internal Medicine, Inje University Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - J E Hwang
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Gwangju, Korea
| | - W K Bae
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Gwangju, Korea
| | - J Cheon
- Department of Hematology and Oncology, Ulsan University Hospital, Ulsan University College of Medicine, Ulsan, Korea
| | - J O Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University College of Medicine, Seoul, Korea
| | - J Y Hong
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University College of Medicine, Seoul, Korea
| | - J H Kang
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
| | - J H Kim
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
| | - S H Lim
- Division of Hematology-Oncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - J W Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - J-W Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - C Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - H J Choi
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
34
|
Conley BA, Staudt L, Takebe N, Wheeler DA, Wang L, Cardenas MF, Korchina V, Zenklusen JC, McShane LM, Tricoli JV, Williams PM, Lubensky I, O’Sullivan-Coyne G, Kohn E, Little RF, White J, Malik S, Harris LN, Mann B, Weil C, Tarnuzzer R, Karlovich C, Rodgers B, Shankar L, Jacobs PM, Nolan T, Berryman SM, Gastier-Foster J, Bowen J, Leraas K, Shen H, Laird PW, Esteller M, Miller V, Johnson A, Edmondson EF, Giordano TJ, Kim B, Ivy SP. The Exceptional Responders Initiative: Feasibility of a National Cancer Institute Pilot Study. J Natl Cancer Inst 2021; 113:27-37. [PMID: 32339229 PMCID: PMC7781457 DOI: 10.1093/jnci/djaa061] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 02/27/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Tumor molecular profiling from patients experiencing exceptional responses to systemic therapy may provide insights into cancer biology and improve treatment tailoring. This pilot study evaluates the feasibility of identifying exceptional responders retrospectively, obtaining pre-exceptional response treatment tumor tissues, and analyzing them with state-of-the-art molecular analysis tools to identify potential molecular explanations for responses. METHODS Exceptional response was defined as partial (PR) or complete (CR) response to a systemic treatment with population PR or CR rate less than 10% or an unusually long response (eg, duration >3 times published median). Cases proposed by patients' clinicians were reviewed by clinical and translational experts. Tumor and normal tissue (if possible) were profiled with whole exome sequencing and, if possible, targeted deep sequencing, RNA sequencing, methylation arrays, and immunohistochemistry. Potential germline mutations were tracked for relevance to disease. RESULTS Cases reflected a variety of tumors and standard and investigational treatments. Of 520 cases, 476 (91.5%) were accepted for further review, and 222 of 476 (46.6%) proposed cases met requirements as exceptional responders. Clinical data were obtained from 168 of 222 cases (75.7%). Tumor was provided from 130 of 168 cases (77.4%). Of 117 of the 130 (90.0%) cases with sufficient nucleic acids, 109 (93.2%) were successfully analyzed; 6 patients had potentially actionable germline mutations. CONCLUSION Exceptional responses occur with standard and investigational treatment. Retrospective identification of exceptional responders, accessioning, and sequencing of pretreatment archived tissue is feasible. Data from molecular analyses of tumors, particularly when combining results from patients who received similar treatments, may elucidate molecular bases for exceptional responses.
Collapse
Affiliation(s)
- Barbara A Conley
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Lou Staudt
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD, USA
| | - Naoko Takebe
- Developmental Therapeutics Clinic, National Cancer Institute, Bethesda, MD, USA
| | - David A Wheeler
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria F Cardenas
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Viktoriya Korchina
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | | | - Lisa M McShane
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - James V Tricoli
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Paul M Williams
- Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Irina Lubensky
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | | | - Elise Kohn
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Richard F Little
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Jeffrey White
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Shakun Malik
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Lyndsay N Harris
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Bhupinder Mann
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Carol Weil
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Roy Tarnuzzer
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD, USA
| | - Chris Karlovich
- Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brian Rodgers
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Lalitha Shankar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Paula M Jacobs
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Tracy Nolan
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sean M Berryman
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Julie Gastier-Foster
- Nationwide Children’s Hospital, Columbus, OH, USA; Van Andel Research Institute, Grand Rapids, MI, USA
| | - Jay Bowen
- Nationwide Children’s Hospital, Columbus, OH, USA; Van Andel Research Institute, Grand Rapids, MI, USA
| | - Kristen Leraas
- Nationwide Children’s Hospital, Columbus, OH, USA; Van Andel Research Institute, Grand Rapids, MI, USA
| | - Hui Shen
- Van Andel Research Institute, Grand Rapids, MI, USA
| | | | - Manel Esteller
- Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Catalonia, Spain
| | | | | | - Elijah F Edmondson
- Pathology and Histology Laboratory, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Benjamin Kim
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - S Percy Ivy
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
35
|
Foschini F, Napolitano F, Servetto A, Marciano R, Mozzillo E, Carratù AC, Santaniello A, De Placido P, Cascetta P, Butturini G, Frigerio I, Regi P, Silvestris N, Delcuratolo S, Vasile E, Vivaldi C, Bianco C, De Placido S, Formisano L, Bianco R. FOLFIRINOX after first-line gemcitabine-based chemotherapy in advanced pancreatic cancer: a retrospective comparison with FOLFOX and FOLFIRI schedules. Ther Adv Med Oncol 2020; 12:1758835920947970. [PMID: 33062062 PMCID: PMC7533956 DOI: 10.1177/1758835920947970] [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: 02/13/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Background Pancreatic adenocarcinoma is the fourth leading cause of cancer-related death. In cases with metastasis, the combination of 5-fluorouracil, irinotecan, and oxaliplatin (FOLFIRINOX) or gemcitabine-based chemotherapy regimens are considered the standard of care. However, the optimal sequence of these regimens is unclear. Methods This retrospective study initially evaluated 186 patients with locally advanced/metastatic pancreatic cancer at three Italian institutions between February 2013 and October 2019. All patients had progressed after receiving gemcitabine-based first-line chemotherapy and were subsequently offered second-line FOLFIRINOX, FOLFOX-6, or FOLFIRI treatment. This study evaluated progression-free survival (PFS), overall survival from the start of second-line treatment (OS2), overall survival from the start of first-line treatment (OS1), and safety outcomes. Results A total of 77 patients received ⩾4 cycles of second-line chemotherapy and were considered eligible: 15 patients received FOLFIRINOX, 32 patients received FOLFOX-6, and 30 patients received FOLFIRI. The FOLFIRINOX group had median PFS of 26.29 weeks and median OS2 of 47.86 weeks, while the FOLFIRI group had median PFS of 10.57 weeks and median OS2 of 25.00 weeks (p = 0.038). No significant differences were observed between the FOLFIRINOX and FOLFOX-6 groups in terms of PFS (26.29 weeks versus 23.07 weeks) or OS2 (47.86 weeks versus 42.00 weeks). The most common grade 3-4 toxicities were anemia, neutropenia, and thrombocytopenia, which occurred more frequently in the FOLFIRINOX and FOLFOX-6 groups. Conclusion Relative to the FOLFIRI regimen, the FOLFIRINOX regimen had a favorable toxicity profile and better survival outcomes. No significant differences were observed relative to the FOLFOX-6 regimen.
Collapse
Affiliation(s)
- Francesca Foschini
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, 80131 Naples, Italy
| | - Fabiana Napolitano
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, 80131 Naples, Italy
| | - Alberto Servetto
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, 80131 Naples, Italy
| | - Roberta Marciano
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, 80131 Naples, Italy
| | - Eleonora Mozzillo
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, 80131 Naples, Italy
| | - Anna Chiara Carratù
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, 80131 Naples, Italy
| | - Antonio Santaniello
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, 80131 Naples, Italy
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, 80131 Naples, Italy
| | - Priscilla Cascetta
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, 80131 Naples, Italy
| | - Giovanni Butturini
- Pancreatic Surgery Unit, Pederzoli Hospital, Peschiera del Garda, Verona, Italy
| | - Isabella Frigerio
- Pancreatic Surgery Unit, Pederzoli Hospital, Peschiera del Garda, Verona, Italy
| | - Paolo Regi
- Pancreatic Surgery Unit, Pederzoli Hospital, Peschiera del Garda, Verona, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II of Bari, Bari, Italy
| | - Sabina Delcuratolo
- Medical Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II of Bari, Bari, Italy
| | - Enrico Vasile
- Unit of Oncology 2, University Hospital of Pisa, Italy
| | | | - Cataldo Bianco
- Department of Experimental and Clinical Medicine, University of Catanzaro Magna Graecia, Catanzaro, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, 80131 Naples, Italy
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, Via Pansini 5, Naples, 80131, Italy
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, Via Pansini 5, Naples, 80131, Italy
| |
Collapse
|
36
|
Wainberg ZA, Feeney K, Lee MA, Muñoz A, Gracián AC, Lonardi S, Ryoo BY, Hung A, Lin Y, Bendell J, Hecht JR. Meta-analysis examining overall survival in patients with pancreatic cancer treated with second-line 5-fluorouracil and oxaliplatin-based therapy after failing first-line gemcitabine-containing therapy: effect of performance status and comparison with other regimens. BMC Cancer 2020; 20:633. [PMID: 32641104 PMCID: PMC7346629 DOI: 10.1186/s12885-020-07110-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/25/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Pancreatic cancer has a poor prognosis and few choices of therapy. For patients with adequate performance status, FOLFIRINOX or gemcitabine plus nab-paclitaxel are preferred first-line treatment. 5-Fluorouracil (5-FU)-based therapy (e.g. FOLFIRI, OFF, or FOLFOX) are often used in patients who previously received gemcitabine-based regimens. A systematic review was conducted of the safety and efficacy of FOLFOX for metastatic pancreatic cancer following prior gemcitabine-based therapy. A Bayesian fixed-effect meta-analysis with adjustment of patient performance status (PS) was conducted to evaluate overall survival (OS) and compare outcomes with nanoliposomal irinotecan combination therapy. METHODS PubMed.gov , FDA.gov , ClinicalTrials.gov , congress abstracts, Cochrane.org library, and EMBASE database searches were conducted to identify randomized controlled trials of advanced/metastatic disease, prior gemcitabine-based therapy, and second-line treatment with 5-FU and oxaliplatin. The database search dates were January 1, 1990-June 30, 2019. Endpoints were OS and severe treatment-related adverse events (TRAEs). Trial-level PS scores were standardized by converting Karnofsky grade scores to Eastern Cooperative Oncology Group (ECOG) Grade, and overall study-weighted PS was calculated based on weighted average of all patients. RESULTS Of 282 studies identified, 11 randomized controlled trials (N = 454) were included in the meta-analysis. Baseline weighted PS scores predicted OS in 10 of the 11 studies, and calculated PS scores of 1.0 were associated with a median OS of 6.3 months (95% posterior interval, 5.4-7.4). After adjusting for baseline PS, FOLFOX had a similar treatment effect profile (median OS, range 2.6-6.7 months) as 5-FU/leucovorin plus nanoliposomal irinotecan therapy (median OS, 6.1 months; 95% confidence interval 4.8-8.9). Neutropenia and fatigue were the most commonly reported Grade 3-4 TRAEs associated with FOLFOX. CONCLUSIONS Baseline PS is a strong prognostic factor when interpreting the efficacy of 5-FU and oxaliplatin-based therapy of pancreatic cancer after progression on first-line gemcitabine-based regimens. When baseline PS is considered, FOLFOX has a similar treatment effect as 5-FU and nanoliposomal irinotecan therapy and a comparable safety profile. These findings suggest that 5-FU and oxaliplatin-based therapies remain an acceptable and alternative second-line treatment option for patients with pancreatic cancer and adequate PS (e.g. ECOG 0-1) following gemcitabine treatment.
Collapse
Affiliation(s)
- Zev A Wainberg
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Kynan Feeney
- Notre Dame University, Fremantle and Edith Cowan University Joondalup, Perth, Australia
| | - Myung Ah Lee
- Catholic University of Korea, Seoul, South Korea
| | - Andrés Muñoz
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Antonio Cubillo Gracián
- HM Universitario Sanchinarro, Centro Integral Oncológico Clara Campal HM-CIOCC, Madrid, Spain.,Departamento de Ciencias Médicas Clínicas Universidad San Pablo CEU, Madrid, Spain
| | - Sara Lonardi
- Istituto Oncologico Veneto - IRCCS, Padova, Italy
| | - Baek-Yeol Ryoo
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Annie Hung
- ARMO Biosciences, a wholly owned subsidiary of Eli Lilly and Company, Redwood City, CA, USA
| | - Yong Lin
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA
| | - J Randolph Hecht
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
37
|
Silvestris N, Brunetti O, Bittoni A, Cataldo I, Corsi D, Crippa S, D’Onofrio M, Fiore M, Giommoni E, Milella M, Pezzilli R, Vasile E, Reni M. Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up of Exocrine Pancreatic Ductal Adenocarcinoma: Evidence Evaluation and Recommendations by the Italian Association of Medical Oncology (AIOM). Cancers (Basel) 2020; 12:E1681. [PMID: 32599886 PMCID: PMC7352458 DOI: 10.3390/cancers12061681] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related death in women (7%) and the sixth in men (5%) in Italy, with a life expectancy of around 5% at 5 years. From 2010, the Italian Association of Medical Oncology (AIOM) developed national guidelines for several cancers. In this report, we report a summary of clinical recommendations of diagnosis, treatment and follow-up of PDAC, which may guide physicians in their current practice. A panel of AIOM experts in upper gastrointestinal cancer malignancies discussed the available scientific evidence supporting the clinical recommendations.
Collapse
Affiliation(s)
- Nicola Silvestris
- Medical Oncology Unit–IRCCS IstitutoTumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (N.S.); (O.B.)
- Department of Biomedical Sciences and Human Oncology-University of Bari Medical School, 70124 Bari, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit–IRCCS IstitutoTumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (N.S.); (O.B.)
| | - Alessandro Bittoni
- Oncology Clinic, AOU Ospedali Riuniti, Polytechnic University of Marche, 60121 Ancona, Italy;
| | - Ivana Cataldo
- Department of Pathology, Hospital Cà Foncello of Treviso, 31100 Treviso, Italy;
| | - Domenico Corsi
- Medical Oncology Unit Azienda Ospedaliera San Giovanni Calibita Fatebene fratelli Roma, 00186 Roma, Italy;
| | - Stefano Crippa
- Division of Pancreatic Surgery, Vita-Salute University, San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Mirko D’Onofrio
- Department of Radiology, G. B. Rossi University Hospital, University of Verona, 37129 Verona, Italy;
| | - Michele Fiore
- Radiation Oncology, Campus Bio-Medico University, 00128 Rome, Italy;
| | - Elisa Giommoni
- Medical Oncology Unit, Department of Oncology and Robotic Surgery, AOU Careggi, 50139 Florence, Italy;
| | - Michele Milella
- Section of Medical Oncology, Department of Medicine, University of Verona and University Hospital Trust, 37129 Verona, Italy;
| | - Raffaele Pezzilli
- Department of Gastroenterology, San Carlo Hospital, 85100 Potenza, Italy;
| | - Enrico Vasile
- Division of Medical Oncology, Pisa University Hospital, 56124 Pisa, Italy;
| | - Michele Reni
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| |
Collapse
|
38
|
De Lisa M, Ballatore Z, Marcantognini G, Pierantoni C, Antognoli S, Pistelli M, Pagliacci A, Berardi R. Irinotecan-Induced Transient Dysarthria: Case Series and Updated Literature Review. Oncol Ther 2020; 8:147-160. [PMID: 32700070 PMCID: PMC7359989 DOI: 10.1007/s40487-019-00106-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Indexed: 12/27/2022] Open
Abstract
Irinotecan-based regimens are used worldwide for the treatment of several recurrent or advanced gastrointestinal malignancies. In this paper we describe the cases of four patients treated in our institution who developed acute dysarthria while receiving intravenous infusion of irinotecan. In all our cases, dysarthria occurred during the infusion of the first course of irinotecan, and then resolved rapidly without any sequelae. Imaging of the brain was performed, but failed to show any evidence of an acute neurological event. We also reviewed the literature on this very uncommon adverse event. The pathogenesis of irinotecan-induced dysarthria is still unknown and is not completely elucidated by the current pharmacodynamic or kinetic explanations; therefore, we could only hypothesize some assumptions.
Collapse
Affiliation(s)
- Mariagrazia De Lisa
- Medical Oncology, Azienda Ospedaliero-Universitaria Ospedali Riuniti-Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Zelmira Ballatore
- Medical Oncology, Azienda Ospedaliero-Universitaria Ospedali Riuniti-Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Giulia Marcantognini
- Medical Oncology, Azienda Ospedaliero-Universitaria Ospedali Riuniti-Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Chiara Pierantoni
- Medical Oncology, Azienda Ospedaliero-Universitaria Ospedali Riuniti-Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Stefania Antognoli
- Medical Oncology, Azienda Ospedaliero-Universitaria Ospedali Riuniti-Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Mirco Pistelli
- Medical Oncology, Azienda Ospedaliero-Universitaria Ospedali Riuniti-Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Alessandra Pagliacci
- Medical Oncology, Azienda Ospedaliero-Universitaria Ospedali Riuniti-Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Rossana Berardi
- Medical Oncology, Azienda Ospedaliero-Universitaria Ospedali Riuniti-Ancona, Università Politecnica delle Marche, Ancona, Italy.
| |
Collapse
|
39
|
Pointet AL, Tougeron D, Pernot S, Pozet A, Béchade D, Trouilloud I, Lourenco N, Hautefeuille V, Locher C, Williet N, Desrame J, Artru P, Soularue E, Le Roy B, Taieb J. Three fluoropyrimidine-based regimens in routine clinical practice after nab-paclitaxel plus gemcitabine for metastatic pancreatic cancer: An AGEO multicenter study. Clin Res Hepatol Gastroenterol 2020; 44:295-301. [PMID: 31607641 DOI: 10.1016/j.clinre.2019.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 08/07/2019] [Accepted: 08/26/2019] [Indexed: 02/04/2023]
Abstract
BACKGROUND A combination of nab-paclitaxel plus gemcitabine (N+G) has recently become a standard first-line treatment in patients with metastatic pancreatic adenocarcinoma (MPA), but there are currently no published data concerning second-line treatment after N+G. The aim of this study was to evaluate the survival outcomes and tolerability of three usual fluoropyrimidine-based regimens FOLFOX, FOLFIRI and FOLFIRINOX after N+G failure in MPA patients. METHODS Patients receiving N+G as first-line regimen were prospectively identified in 11 French centers between January 2014 and January 2017. After disease progression or unacceptable toxicity, patients eligible for second-line therapy were enrolled in the study. The primary endpoint was overall survival following the second-line regimen. Secondary endpoints were objective response, progression-free survival and safety. RESULTS Out of 137 patients treated with N+G as first-line regimen, 61 (44.5%) received second-line chemotherapy, including FOLFOX (39.4%), FOLFIRI (34.4%) or FOLFIRINOX (26.2%). Baseline characteristics were not different between the 3 groups. In particular, median age was 71.7 years, sex ratio was 1/1, and performance status (PS) was 0 in 11.5% of case. Main grade 3 toxicities were neutropenia (4.9%) and nausea (3.3%), without major differences between the groups. No toxic death was observed. Median second-line progression-free survival (PFS) and overall survival (OS) were 2.95 (95% CI: 2.3-5.4) and 5.97 months (95% CI: 4.0-8.0), respectively, with no difference between the 3 groups. Median OS from the start of first-line chemotherapy was 12.7 months (10.4-15.1) and was significantly better in patients receiving FOLFIRI after N+G failure, 18.4 months (95% CI: 11.7-24.1, P<0.05), as compared with FOLFOX or FOLFIRINOX (10.4 and 12.3 months, respectively). CONCLUSION This study suggests that second-line fluoropyrimidine-based regimens after N+G failure are feasible, have a manageable toxicity profile in selected patients with MPA, and are associated with promising clinical outcomes, in particular when combined with irinotecan. Randomized phase 3 trials are needed to confirm this trend.
Collapse
Affiliation(s)
- Anne-Laure Pointet
- Department of Gastroenterology and Digestive Oncology, Georges-Pompidou European Hospital, Assistance publique-Hôpitaux de Paris (AP-HP), Sorbonne Paris Cité Paris Descartes University, Paris, France.
| | - David Tougeron
- Department of Gastroenterology, Poitiers University Hospital, Poitiers, France
| | - Simon Pernot
- Department of Gastroenterology and Digestive Oncology, Georges-Pompidou European Hospital, Assistance publique-Hôpitaux de Paris (AP-HP), Sorbonne Paris Cité Paris Descartes University, Paris, France
| | - Astrid Pozet
- Methodology and quality of life in oncology unit, (Inserm UMR 1098), Besançon university Hospital, Besançon, France
| | - Dominique Béchade
- Department of Medical Oncology, Anticancer Center Bergonié Institute, Bordeaux University, Bordeaux, France
| | - Isabelle Trouilloud
- Department of Gastroenterology and Digestive Oncology, Saint-Antoine Hospital, Assistance publique-Hôpitaux de Paris (AP-HP), Pierre et Marie Curie University, Paris, France
| | - Nelson Lourenco
- Department of Gastroenterology, Saint-Louis Hospital, Assistance publique-Hôpitaux de Paris (AP-HP), Paris VII University, Paris, France
| | | | | | - Nicolas Williet
- Department of Gastroenterology, Saint-Étienne University Hospital, Saint-Étienne, France
| | | | | | - Emilie Soularue
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance publique-Hôpitaux de Paris (AP-HP), Paris Sud University, Le Kremlin Bicêtre, France
| | - Bertrand Le Roy
- Department of Digestive surgery and oncology, Clermont-Ferrand University Hospital, France
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, Georges-Pompidou European Hospital, Assistance publique-Hôpitaux de Paris (AP-HP), Sorbonne Paris Cité Paris Descartes University, Paris, France
| |
Collapse
|
40
|
Chae H, Jeong H, Cheon J, Chon HJ, Ryu H, Kim IH, Kang MJ, Jeong JH, Ryoo BY, Kim KP, Yoo C. Efficacy and safety of second-line nab-paclitaxel plus gemcitabine after progression on FOLFIRINOX for unresectable or metastatic pancreatic ductal adenocarcinoma: multicenter retrospective analysis. Ther Adv Med Oncol 2020; 12:1758835920923424. [PMID: 32523632 PMCID: PMC7257856 DOI: 10.1177/1758835920923424] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/03/2020] [Indexed: 12/12/2022] Open
Abstract
Background: FOLFIRINOX (fluorouracil, folinic acid, irinotecan plus oxaliplatin) is an
effective standard first-line treatment option for advanced pancreatic
ductal adenocarcinoma (PDAC). There is no clear consensus on the second-line
treatment following progression on FOLFIRINOX. In this multicenter
retrospective analysis, we evaluated the efficacy and tolerability of
second-line nab-P/Gem (nab-paclitaxel and gemcitabine) after progression on
FOLFIRNOX in PDAC. Methods: Patients with unresectable or metastatic PDAC who received nab-P/Gem after
progression on FOLFIRINOX between February 2016 and February 2019 were
identified from five referral cancer centers in South Korea. Baseline
characteristics, treatment history, survival outcomes, and toxicity profile
were obtained retrospectively from medical records. Results: A total of 102 patients treated with second-line nab-P/Gem for advanced PDAC
after progression on FOLFIRINOX were included. At the time of nab-P/Gem, the
median age was 60 years, with males comprising 49.0%, and most (75.5%) had
metastatic disease. Patients received a median of three cycles (range 1–12)
of nab-P/Gem. The median overall survival (OS) and progression-free survival
(PFS) from the start of second-line nab-P/Gem therapy were 9.8 (95% CI,
8.9–10.6) and 4.6 months (3.7–5.5), respectively. A partial response was
achieved in 8.5%, and the disease control rate was 73.6%. From the start of
first-line FOLFIRIOX, the OS1+2 and PFS1+2 were 20.9
(15.7–26.1) and 13.9 (10.8–17.0) months, respectively, with a 2-year
survival rate of 45.1%. There was no treatment-related mortality and grade
⩾3 toxicity was observed in 60.2%. Conclusion: Our results showed that nab-P/Gem was an effective and tolerable second-line
treatment option in medically fit patients with advanced PDAC who progressed
on first-line FOLFIRNOX. ClinicalTrials.gov identifier: NCT04133155
Collapse
Affiliation(s)
- Heejung Chae
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, South Korea
| | - Hyehyun Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, South Korea
| | - Jaekyung Cheon
- Division of Hematology and Oncology, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, South Korea
| | - Hong Jae Chon
- Department of Medical Oncology, CHA Bundang Medical Center, CHA University, South Korea
| | - Hyewon Ryu
- Division of Hematology and Oncology, Department of Internal Medicine, Chungnam National University Hospital, Chungnam National University College of Medicine, South Korea
| | - Il-Hwan Kim
- Department of Oncology, Haeundae Paik Hospital, Cancer Center, Inje University College of Medicine, Busan, South Korea
| | - Myoung Joo Kang
- Department of Oncology, Haeundae Paik Hospital, Cancer Center, Inje University College of Medicine, Busan, South Korea
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, South Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, South Korea
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| |
Collapse
|
41
|
Bang Y, Li C, Lee K, Chiu C, Park JO, Shan Y, Kim JS, Chen J, Shim H, Rau K, Choi HJ, Oh D, Belanger B, Chen L. Liposomal irinotecan in metastatic pancreatic adenocarcinoma in Asian patients: Subgroup analysis of the NAPOLI-1 study. Cancer Sci 2020; 111:513-527. [PMID: 31789476 PMCID: PMC7004519 DOI: 10.1111/cas.14264] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/11/2019] [Accepted: 11/19/2019] [Indexed: 12/18/2022] Open
Abstract
The global, randomized NAPOLI-1 phase 3 trial reported a survival benefit with liposomal irinotecan (nal-IRI) plus 5-fluorouracil/leucovorin (nal-IRI+5-FU/LV) in patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) after previous gemcitabine-based therapy. Median overall survival (OS) with nal-IRI+5-FU/LV was 6.1 vs 4.2 months with 5-FU/LV alone (unstratified hazard ratio [HR] = 0.67, P = .012). Herein, we report efficacy and safety results from a post-hoc subgroup analysis of Asian patients treated at Asian centers. Primary study endpoint was OS; secondary endpoints included progression-free survival (PFS), objective response rate (ORR), and safety. Patients receiving nal-IRI+5-FU/LV (n = 34) had significantly longer median OS versus 5-FU/LV (n = 35) (8.9 vs 3.7 months; unstratified HR = 0.51, P = .025). Patients had significantly increased median PFS with nal-IRI+5-FU/LV versus 5-FU/LV (4.0 vs 1.4; unstratified HR = 0.48, P = .011), and increased ORR (8.8% vs 0; P = .114). nal-IRI monotherapy (n = 50) numerically improved efficacy endpoints versus 5-FU/LV (n = 48): median OS was 5.8 versus 4.3 months (HR = 0.83, P = .423) and median PFS was 2.8 versus 1.4 months (HR = 0.69, P = .155). Grade ≥3 neutropenia was reported more frequently with nal-IRI+5-FU/LV versus 5-FU/LV (54.5% vs 3.4%), and incidence of grade ≥3 diarrhea was comparable between the two arms (3.0% vs 6.9%). This subgroup analysis confirms nal-IRI+5-FU/LV as an efficacious treatment option that improves survival in Asian patients with mPDAC that progressed after gemcitabine-based therapy, with a safety profile agreeing with previous findings. The nal-IRI+5-FU/LV regimen should represent a new standard of care for these patients in Asia. (Clinicaltrials.gov: NCT01494506).
Collapse
Affiliation(s)
- Yung‐Jue Bang
- Seoul National University College of MedicineSeoulKorea
| | - Chung‐Pin Li
- Division of Gastroenterology and HepatologyDepartment of MedicineTaipei Veterans General HospitalTaipeiTaiwan
- National Yang‐Ming University School of MedicineTaipeiTaiwan
| | - Kyung‐Hun Lee
- Seoul National University HospitalCancer Research InstituteSeoul National University College of MedicineSeoulKorea
| | | | - Joon Oh Park
- Samsung Medical CenterSungkyunkwan University School of MedicineSeoulKorea
| | - Yan‐Shen Shan
- Department of SurgeryNational Cheng Kung University HospitalTainanTaiwan
| | | | - Jen‐Shi Chen
- Department of Hematology‐OncologyLinkou Chang Gung Memorial Hospital and Chang Gung UniversityTao‐YuanTaiwan
| | | | - Kun‐Ming Rau
- Kaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan
| | | | - Do‐Youn Oh
- Seoul National University HospitalCancer Research InstituteSeoul National University College of MedicineSeoulKorea
| | | | - Li‐Tzong Chen
- National Institute of Cancer ResearchNational Health Research Institutes (NHRI)TainanTaiwan
- Department of Internal MedicineNational Cheng Kung University HospitalNational Cheng Kung UniversityTainanTaiwan
- Department of Internal MedicineKaohsiung Medical University HospitalKaohsiung Medical UniversityKaohsiungTaiwan
| |
Collapse
|
42
|
Gupta M, Iyer R, Fountzilas C. Poly(ADP-Ribose) Polymerase Inhibitors in Pancreatic Cancer: A New Treatment Paradigms and Future Implications. Cancers (Basel) 2019; 11:E1980. [PMID: 31835379 PMCID: PMC6966572 DOI: 10.3390/cancers11121980] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/30/2019] [Accepted: 12/06/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy. Most of the patients of PDAC present at later stages of disease and have a five-year survival rate of less than 10%. About 5-10% PDAC cases are hereditary in nature and have DNA damage repair (DDR) mutations such as BRCA 1 and 2. Besides having implications on screening and prevention strategies, these mutations can confer sensitivity to platinum-based therapies and determine eligibility for poly(ADP-ribose) polymerase inhibitors (PARPi). In the presence of DDR mutations and PARPi, the cells are unable to utilize the error-free process of homologous recombination repair, leading to accumulation of double stranded DNA breaks and cell death eventually. Various PARPi are in clinical development in PDAC in different subgroup of patients as monotherapies and in combination with other therapeutics. This review would focus on the mechanism of action of PARPi, history of development in PDAC, resistance mechanisms and future directions.
Collapse
Affiliation(s)
- Medhavi Gupta
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Renuka Iyer
- Department of Medicine/Division of GI Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Christos Fountzilas
- Department of Medicine/Division of GI Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| |
Collapse
|
43
|
Lambert A, Schwarz L, Borbath I, Henry A, Van Laethem JL, Malka D, Ducreux M, Conroy T. An update on treatment options for pancreatic adenocarcinoma. Ther Adv Med Oncol 2019; 11:1758835919875568. [PMID: 31598142 PMCID: PMC6763942 DOI: 10.1177/1758835919875568] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 08/19/2019] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is one of the most lethal solid organ tumors. Due to the rising incidence, late diagnosis, and limited treatment options, it is expected to be the second leading cause of cancer deaths in high income countries in the next decade. The multidisciplinary treatment of this disease depends on the stage of cancer at diagnosis (resectable, borderline, locally advanced, and metastatic disease), and combines surgery, chemotherapy, chemoradiotherapy, and supportive care. The landscape of multidisciplinary pancreatic cancer treatment is changing rapidly, especially in locally advanced disease, and the number of treatment options in metastatic disease, including personalized medicine, innovative targets, immunotherapy, therapeutic vaccines, adoptive T-cell transfer, or stemness inhibitors, will probably expand in the near future. This review summarizes the current literature and provides an overview of how new therapies or new therapeutic strategies (neoadjuvant therapies, conversion surgery) will guide multidisciplinary disease management, future clinical trials, and, hopefully, will increase overall survival.
Collapse
Affiliation(s)
- Aurélien Lambert
- Department of Medical Oncology, Institut de Cancérologie de Lorraine and Université de Lorraine, Nancy, France
| | - Lilian Schwarz
- Department of Digestive Surgery, Rouen University Hospital and Université de Rouen Normandie, France
| | - Ivan Borbath
- Department of Gastroenterology and Digestive Oncology, Cliniques Universitaires Saint-Luc and Université Catholique de Louvain, Brussels, Belgium
| | - Aline Henry
- Department of Supportive Care in Oncology, Institut de Cancérologie de Lorraine, Nancy, France
| | - Jean-Luc Van Laethem
- Department of Gastroenterology and Digestive Oncology, Erasme University Hospital, Université Libre de Bruxelles, Belgium
| | - David Malka
- Department of Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Michel Ducreux
- Department of Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Thierry Conroy
- Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 50519 Vandoeuvre-lès-Nancy CEDEX, France
| |
Collapse
|
44
|
Tossey JC, Reardon J, VanDeusen JB, Noonan AM, Porter K, Arango MJ. Comparison of conventional versus liposomal irinotecan in combination with fluorouracil for advanced pancreatic cancer: a single-institution experience. Med Oncol 2019; 36:87. [PMID: 31494781 DOI: 10.1007/s12032-019-1309-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/27/2019] [Indexed: 12/15/2022]
Abstract
The majority of pancreatic cancers are diagnosed at an advanced stage, when surgical options are limited and treatment relies on systemic chemotherapy. In the NAPOLI-1 trial, liposomal irinotecan in combination with fluorouracil (nal-iri/5FU) was shown to improve overall survival when compared to fluorouracil alone for metastatic pancreatic cancer. Other retrospective studies have shown the combination of fluorouracil and conventional irinotecan (FOLFIRI) to be a viable option, though no randomized trials have compared nal-iri/5FU to FOLFIRI. The purpose of this single-center, retrospective, cohort study was to determine if nal-iri/5FU and FOLFIRI are similarly effective for the treatment of advanced pancreatic cancer. Due to the potential for treatment bias, inverse probability of treatment weighting was utilized to correct for baseline differences between the groups. The primary outcome of progression-free survival was similar at 4.1 months for nal-iri/5FU and 3.1 months for FOLFIRI. Overall survival and adverse effect frequency were also similar. Pegfilgrastim was used in 16% and 15% of patients, respectively, and nal-iri/5FU patients required significantly less atropine during treatment (36 vs. 70%). A cost analysis was conducted and concluded that the treatment with nal-iri/5FU was nearly 30 times more expensive than FOLFIRI treatment. Together, these data suggest a potential role for FOLFIRI for the treatment of advanced pancreatic cancer in the absence of clear benefits in effectiveness, toxicity, or cost for nal-iri/5FU.
Collapse
Affiliation(s)
- Justin C Tossey
- Department of Pharmacy, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, 460 West 10th Avenue, Columbus, OH, 43210, USA
| | - Joshua Reardon
- Department of Pharmacy, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, 460 West 10th Avenue, Columbus, OH, 43210, USA
| | - Jeffrey B VanDeusen
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, 460 West 10th Avenue, Columbus, OH, USA
| | - Anne M Noonan
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, 460 West 10th Avenue, Columbus, OH, USA
| | - Kyle Porter
- Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University, 1800 Cannon Drive, Columbus, OH, USA
| | - Matthew J Arango
- Department of Pharmacy, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, 460 West 10th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
45
|
Kieler M, Unseld M, Bianconi D, Scheithauer W, Prager GW. A real-world analysis of second-line treatment options in pancreatic cancer: liposomal-irinotecan plus 5-fluorouracil and folinic acid. Ther Adv Med Oncol 2019; 11:1758835919853196. [PMID: 31360237 PMCID: PMC6640066 DOI: 10.1177/1758835919853196] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 04/12/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Nanoliposomal irinotecan (nal-IRI) plus 5-fluorouracil/leucovorin (5-FU/LV) is a novel treatment option for gemcitabine-pretreated metastatic pancreatic adenocarcinoma (PAC) patients, but real-world evidence is rare. Our aim was to determine the effectiveness and tolerability of this regimen in advanced PAC patients and to compare it with oxaliplatin plus fluoropyrimidines in the second-line setting after failure of gemcitabine. Methods: This is a retrospective single-center analysis of all patients who have been treated with nal-IRI plus 5-FU/LV. To compare its effectiveness with other second-line treatment options, all patients who had received oxaliplatin plus fluoropyrimidines after gemcitabine-based chemotherapy were also eligible for analysis. Results: Fifty-two patients were treated with nal-IRI plus 5-FU/LV between April 2016 and August 2018. The median progression-free survival (PFS) was 3.84 months and the median overall survival (OS) was 6.79 months. Median OS from the beginning of the treatment for advanced disease was 19.9 months. Median PFS in patients that received nal-IRI plus 5-FU/LV as second-line treatment after gemcitabine-based chemotherapy was 4.49 months whereas median PFS in a matched cohort of patients treated with oxaliplatin plus fluoropyrimidines was 3.44 months (p = 0.007). Between these two groups the median OS of patients with CA 19-9 levels above the statistical median (⩾772.8 kU/l) differed significantly (9.33 versus 6.18 months, p = 0.038). Conclusion: Our data confirms the effectiveness of nal-IRI plus 5-FU/LV treatment as a well-tolerated regimen in the treatment of advanced PAC and extends available data on its use as a second-line treatment option when compared with oxaliplatin plus fluoropyrimidines.
Collapse
Affiliation(s)
- Markus Kieler
- Department of Medicine I, Division of Oncology, Comprehensive Cancer Center, Medical University Vienna, Austria
| | - Matthias Unseld
- Department of Medicine I, Division of Oncology, Comprehensive Cancer Center, Medical University Vienna, Austria
| | - Daniela Bianconi
- Department of Medicine I, Division of Oncology, Comprehensive Cancer Center, Medical University Vienna, Austria
| | - Werner Scheithauer
- Department of Medicine I, Division of Oncology, Comprehensive Cancer Center, Medical University Vienna, Austria
| | - Gerald W Prager
- Department of Medicine I, Division of Oncology, Comprehensive Cancer Center, Medical University Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
46
|
Lee K, Bang K, Yoo C, Hwang I, Jeong JH, Chang HM, Oh D, Song TJ, Park DH, Lee SS, Lee SK, Kim MH, Park JH, Kim KP, Ryoo BY. Clinical Outcomes of Second-Line Chemotherapy after Progression on Nab-Paclitaxel Plus Gemcitabine in Patients with Metastatic Pancreatic Adenocarcinoma. Cancer Res Treat 2019; 52:254-262. [PMID: 31291709 PMCID: PMC6962478 DOI: 10.4143/crt.2019.190] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/07/2019] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Since the introduction of nab-paclitaxel plus gemcitabine (nab-P+GEM) as first-line (1L) treatment for metastatic pancreatic adenocarcinoma (mPDAC), optimal second-line (2L) chemotherapy after progression is unclear. We assessed clinical outcomes of 2L chemotherapy for disease that progressed on 1L nab-P+GEM. Materials and Methods Among the 203 patients previously treated with 1L nab-P+GEM for mPDAC at Asan Medical Center, between February and December 2016, records of 120 patients receiving 2L chemotherapy after progression on nab-P+GEM were retrospectively reviewed. The response rate and survival were evaluated along with analysis of prognostic factors. RESULTS Fluoropyrimidine-oxaliplatin doublets (FOLFOX or XELOX) were used in 78 patients (65.0%), fluoropyrimidine monotherapy in 37 (30.8%), and liposomal irinotecan plus fluorouracil in two (1.7%). The median progression-free survival (PFS) and overall survival (OS) were 3.29 months and 7.33 months from the start of 2L therapy. Fluoropyrimidine-oxaliplatin regimens and fluoropyrimidine monotherapy did not yield significantly different median PFS (2.89 months vs. 3.81 months, p=0.40) or OS (7.04 months vs. 7.43 months, p=0.86). A high neutrophil-lymphocyte ratio (> 2.2) and a short time to progression with 1L nab-P+GEM (< 6.4 months) were independent prognostic factors of poor OS with 2L therapy. CONCLUSION 2L fluoropyrimidine-oxaliplatin doublets and fluoropyrimidine monotherapy after failure of 1L nab-P+GEM had modest efficacy, with no differences in treatment outcomes between them. Further investigation is warranted for the optimal 2L chemo-regimens and sequencing of systemic chemotherapy for patients with mPDAC.
Collapse
Affiliation(s)
- Kyoungmin Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyunghye Bang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Inhwan Hwang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Heung-Moon Chang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dongwook Oh
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Tae Jun Song
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Do Hyun Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang Soo Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung Koo Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Myung-Hwan Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin-Hong Park
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
47
|
Tesfaye AA, Wang H, Hartley ML, He AR, Weiner L, Gabelia N, Kapanadze L, Shezad M, Brody JR, Marshall JL, Pishvaian MJ. A Pilot Trial of Molecularly Tailored Therapy for Patients with Metastatic Pancreatic Ductal Adenocarcinoma. J Pancreat Cancer 2019; 5:12-21. [PMID: 31065624 PMCID: PMC6503449 DOI: 10.1089/pancan.2019.0003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose: Despite the wide adoption of tumor molecular profiling, there is a dearth of evidence linking molecular biomarkers for treatment selection to prediction of treatment outcomes in patients with metastatic pancreatic cancer. We initiated a pilot study to test the feasibility of designing a larger phase II trial of molecularly tailored treatment for metastatic pancreatic cancer. Methods: Our study aimed to assess the feasibility of following a treatment algorithm based on the expression of three published predictive markers of response to chemotherapy: ribonucleotide reductase catalytic subunit M1 (for gemcitabine); excision repair cross-complementation group 1 (for platinum agents); and thymidylate synthase (for 5-fluorouracil) in patients with untreated, metastatic pancreatic cancer. Results of the tumor biopsy analysis were used to assign patients to one of seven doublet regimens. Key secondary objectives included response rate (RR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). Results: Between December 2012 and March 2015, 30 patients were enrolled into the study. Ten patients failed screening primarily due to inadequate tumor tissue availability. Of the remaining 20 patients, 19 were assigned into 6 different chemotherapy doublets, and achieved an RR of 28%, with a DCR rate of 78%. The median PFS and OS were 5.78 and 8.21 months, respectively. Conclusions: The incorporation of biomarkers into a treatment algorithm is feasible and resulted in a PFS and OS similar to other doublet therapies for patients with metastatic pancreatic cancer. Based on the results from this pilot study, a larger phase II randomized trial of molecularly targeted therapy versus physicians' choice of standard of care has been initiated in the second-line setting (NCT02967770).
Collapse
Affiliation(s)
- Anteneh A Tesfaye
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Hongkun Wang
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Marion L Hartley
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Aiwu Ruth He
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Louis Weiner
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Nina Gabelia
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Lana Kapanadze
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Muhammad Shezad
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Jonathan R Brody
- Department of Surgery, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - John L Marshall
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Michael J Pishvaian
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| |
Collapse
|
48
|
Woo W, Carey ET, Choi M. Spotlight on liposomal irinotecan for metastatic pancreatic cancer: patient selection and perspectives. Onco Targets Ther 2019; 12:1455-1463. [PMID: 30863113 PMCID: PMC6391121 DOI: 10.2147/ott.s167590] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is a highly lethal disease, where the mortality closely matches increasing incidence. Pancreatic ductal adenocarcinoma (PDAC) is the most common histologic type that tends to metastasize early in tumor progression. For metastatic PDAC, gemcitabine had been the mainstay treatment for the past three decades. The treatment landscape has changed since 2010, and current first-line chemotherapy includes triplet drugs like FOLFIRINOX (folinic acid, 5-fluorouracil, irinotecan, and oxaliplatin), and doublet agents like nab-paclitaxel and gemcitabine. Nanoliposomal encapsulated irinotecan (nal-IRI) was developed as a novel formulation to improve drug delivery, effectiveness, and limit toxicities. Nal-IRI, in combination with leucovorin-modulated fluorouracil (5-FU/LV), was found in a large randomized phase III clinical trial (NAPOLI-1) to significantly improve overall survival in patients who progressed on gemcitabine-based therapy. This review will focus on the value of using nal-IRI, toxicities, recent clinical experiences, and tools to improve patient outcomes in this setting.
Collapse
Affiliation(s)
- Wonhee Woo
- Division of Hematology/Oncology, Department of Medicine, Stony Brook University, Stony Brook, NY, USA,
| | - Edward T Carey
- Division of Hematology/Oncology, Department of Medicine, Stony Brook University, Stony Brook, NY, USA,
| | - Minsig Choi
- Division of Hematology/Oncology, Department of Medicine, Stony Brook University, Stony Brook, NY, USA,
| |
Collapse
|
49
|
Chung MJ, Kang H, Kim HG, Hyun JJ, Lee JK, Lee KH, Noh MH, Kang DH, Lee SH, Bang S, Pancreatobiliary Cancer Study Group of Korean Society of Gastrointestinal Cancer. Multicenter phase II trial of modified FOLFIRINOX in gemcitabine-refractory pancreatic cancer. World J Gastrointest Oncol 2018; 10:505-515. [PMID: 30595804 PMCID: PMC6304301 DOI: 10.4251/wjgo.v10.i12.505] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/24/2018] [Accepted: 11/25/2018] [Indexed: 02/05/2023] Open
Abstract
AIM To evaluate the efficacy and safety of modified FOLFIRINOX as a second-line treatment for gemcitabine (GEM)-refractory unresectable pancreatic cancer (PC). METHODS This study was a prospective, multicenter, one-arm, open-label, phase II trial. Patients with unresectable PC, who showed disease progression during GEM-based chemotherapy were enrolled. All patients were administered FOLFIRINOX with reduced irinotecan and oxaliplatin (RIO; irinotecan 120 mg/m2 and oxaliplatin 60 mg/m2), which was set according to the phase I study of FOLFIRINOX. The objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), adverse events were evaluated. Additionally, changes in quality of life (QoL) were assessed using a questionnaire on QoL. RESULTS Between August 2015 and May 2016, a total of 48 patients were enrolled. The median follow-up time was 259 d with a median of 8.5 cycles. The ORR and DCR were 18.8% and 62.5%, respectively, including one patient who showed complete remission. The median PFS was 5.8 mo [95% confidence interval (CI): 3.7-7.9] and median OS was 9.0 mo (95%CI: 6.4-11.6). Neutropenia (64.6%) was the most common grade 3-4 adverse event, followed by febrile neutropenia (16.7%). Although 14.6% of patients experienced grade 3 fatigue, most non-hematologic AEs were under grade 2. In the QoL analysis, the global health status score before treatment was not different from the score at the last visit after treatment (45.43 ± 22.88 vs 48.66 ± 24.14, P = 0.548). CONCLUSION FOLFIRINOX with RIO showed acceptable toxicity and promising efficacy for GEM-refractory unresectable PC. However, this treatment requires careful observation of treatment-related hematologic toxicities.
Collapse
Affiliation(s)
- Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Huapyong Kang
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Ho Gak Kim
- Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu 42471, South Korea
| | - Jong Jin Hyun
- Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan 15355, South Korea
| | - Jun Kyu Lee
- Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang 10326, South Korea
| | - Kwang Hyuck Lee
- Division of Gastroenterology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Myung Hwan Noh
- Department of Internal Medicine, Dong-A University College of Medicine, Busan 49201, South Korea
| | - Dae Hwan Kang
- Department of Internal Medicine, Medical Research Institute, Pusan National University School of Medicine and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, South Korea
| | - Sang Hyub Lee
- Departments of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Seungmin Bang
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, South Korea
| | | |
Collapse
|
50
|
Neuzillet C, Gaujoux S, Williet N, Bachet JB, Bauguion L, Colson Durand L, Conroy T, Dahan L, Gilabert M, Huguet F, Marthey L, Meilleroux J, de Mestier L, Napoléon B, Portales F, Sa Cunha A, Schwarz L, Taieb J, Chibaudel B, Bouché O, Hammel P. Pancreatic cancer: French clinical practice guidelines for diagnosis, treatment and follow-up (SNFGE, FFCD, GERCOR, UNICANCER, SFCD, SFED, SFRO, ACHBT, AFC). Dig Liver Dis 2018; 50:1257-1271. [PMID: 30219670 DOI: 10.1016/j.dld.2018.08.008] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND This document is a summary of the French intergroup guidelines regarding the management of pancreatic adenocarcinoma (PA), updated in July 2018. DESIGN This collaborative work was produced under the auspices of all French medical and surgical societies involved in the management of PA. It is based on the previous guidelines, recent literature review and expert opinions. Recommendations were graded in three categories, according to the level of evidence. RESULTS Over the last seven years, significant changes in PA management have been implemented in clinical practice. Imaging/staging: diffusion magnetic resonance imaging is useful before surgery to rule out small liver metastases. SURGERY centralization of pancreatic surgery in expert centers is associated with a decreased postoperative mortality. Adjuvant chemotherapy: modified FOLFIRINOX in fit patients, or gemcitabine, or 5-FU, or gemcitabine plus capecitabine, to be discussed on a case-by-case basis. Locally advanced PA: no survival benefit of chemoradiotherapy. Metastatic PA: FOLFIRINOX and gemcitabine plus nab-paclitaxel combination are first-line standards in fit patients; second-line with 5FU/nal-IRI or 5FU/oxaliplatin combination after first-line gemcitabine. CONCLUSION Guidelines for management of PA are continuously evolving and need to be regularly updated. This constant progress is made possible through clinical and translational research. However, as each individual case is particular, they cannot substitute to multidisciplinary tumor board discussion.
Collapse
Affiliation(s)
- Cindy Neuzillet
- Department of Medical Oncology, Curie Institute, Versailles Saint-Quentin University (UVSQ), Saint-Cloud, France.
| | - Sébastien Gaujoux
- Department of Digestive, Hepato-Biliary and Pancreatic Surgery, Cochin Hospital, AP-HP, Paris Descartes Faculty of Medicine, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Nicolas Williet
- Hepato-Gastroenterology Department, University Hospital of Saint-Etienne, Saint Priest en Jarez, France
| | - Jean-Baptiste Bachet
- Hepato-Gastroenterology Department, Pitié Salpétrière University Hospital, AP-HP, Paris Cedex 13, France
| | - Lucile Bauguion
- Hepato-Gastroenterology Department, Departmental Hospital Center, La Roche sur Yon, France
| | - Laurianne Colson Durand
- Department of Radiotherapy, Henri Mondor Hospital, AP-HP, Université Paris Est Creteil, Créteil, France
| | - Thierry Conroy
- Department of Medical Oncology, Lorraine Institute of Oncology and Lorraine University, Vandoeuvre-lès-Nancy Cedex, France
| | - Laetitia Dahan
- Digestive Oncology Department, "DACCORD" (Digestif, Anatomie pathologique, Chirurgie, CISIH, Oncologie, Radiothérapie, Dermatologie) pole, CHU Timone, Marseille Cedex 05, France
| | - Marine Gilabert
- Paoli Calmettes Institute, Department of Medical Oncology and Cancer Research Center of Marseille (CRCM), INSERM U1068 Stress Cell, Aix-Marseille University, Marseille, France
| | - Florence Huguet
- Department of Oncology and Radiotherapy, Tenon Hospital, East Paris University Hospitals, AP-HP, Paris Sorbonne University, Paris, France
| | - Lysiane Marthey
- Gastroenterology Department, Béclère Hospital, AP-HP, Clamart, France
| | - Julie Meilleroux
- Pathology Department, Toulouse University Hospital, Toulouse, France
| | - Louis de Mestier
- Department of Gastroenterology-Pancreatology, Beaujon Hospital, APHP, Paris 7 University, Clichy, France
| | - Bertrand Napoléon
- Jean Mermoz Private Hospital, Ramsay Générale de Santé, Lyon, France
| | - Fabienne Portales
- Digestive Oncology Department, Regional Institute of Cancer, Montpellier, France
| | - Antonio Sa Cunha
- INSERM UMR 935, Paul Brousse Hospital, Hepatobiliary Center, AP-HP, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Lilian Schwarz
- Department of Digestive Surgery, Hôpital Charles Nicolle, Rouen University Hospital, Rouen, France and Genomic and Personalized Medicine in Cancer and Neurological Disorders, UMR 1245 INSERM, Rouen University, France
| | - Julien Taieb
- Hepato-Gastroenterology and Digestive Oncology Department, Georges Pompidou European Hospital, AP-HP, Paris, France
| | - Benoist Chibaudel
- Department of Medical Oncology, Franco-British Institute, Levallois-Perret, France
| | - Olivier Bouché
- Hepato-Gastroenterology and Digestive Oncology Department, Robert Debré University Hospital, Avenue Général Koenig, 51092 Reims Cedex, France
| | - Pascal Hammel
- Department of Digestive Oncology, Beaujon University Hospital (AP-HP), Paris VII Diderot University, Clichy-la-Garenne, France.
| |
Collapse
|