601
|
Abrini H, Amzerin M, El Baaboua A, Aboulaghras S, Bouhda A, El Mrabet FZ. Comparison of different Pancreatic cancer treatments: a three-year retrospective study in the oncology center of Tangier university hospital, Morocco. BMC Gastroenterol 2023; 23:452. [PMID: 38129797 PMCID: PMC10734188 DOI: 10.1186/s12876-023-03071-0] [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: 07/23/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Pancreatic cancer is among the most lethal malignancies, with a 5-year overall survival (OS) of less than 10% for all stages. The present study aims to evaluate the epidemiological and clinical characteristics, as well as the results of different treatments of patients diagnosed and treated between 2019 and 2021 in the Oncology Center of Tangier, University Hospital, Morocco. METHODS To compare the evolution of the pancreatic cancer between the different chemotherapy regimens, a retrospective study was performed using data collected over a period of 3 years. For each patient, the data were described and statistically analyzed in the dedicated operating sheet. RESULTS 55 pancreatic cancer patients were included in this study, and the median follow up was 3 months. The mean age of patients was 59.5 ± 10.3 years (extremes 34-79) and the sex ratio male/female was 0.9. Most patients were diagnosed with adenocarcinoma (92.3%), but metastatic stage was the most frequent (56.4%). The surgery was applied to 16.36% of patients. 10.9% of patients have received adjuvant chemotherapy and 76.4% received palliative chemotherapy. Chemotherapy regimens included mainly Gemcitabine and Folfirinox. The median OS was significantly longer for patients treated with Folfirinox versus Gemcitabine (6 months versus 3 months, p-value < 0.016). The median OS for patients that received Folfirinox and Gemcitabine successively (19.7 months) was significantly longer compared to patients that received a monotherapy with either Folfirinox or Gemcitabine alone (p-value < 0.016). CONCLUSION These findings reinforce the use of advanced methods for earlier detection of pancreatic cancer and the development of effective immunotherapies or more targeted therapies.
Collapse
Affiliation(s)
- Houda Abrini
- Department of Medical Oncology, Faculty of Medicine and Pharmacy, Ahmad Bin Zayed Al Nahyan Center of Cancer Treatment, Mohammed VI University Hospital of Tangier, Abdelmalek Essaadi University, Tangier, Morocco.
| | - Mounia Amzerin
- Department of Medical Oncology, Faculty of Medicine and Pharmacy, Ahmad Bin Zayed Al Nahyan Center of Cancer Treatment, Mohammed VI University Hospital of Tangier, Abdelmalek Essaadi University, Tangier, Morocco
| | - Aicha El Baaboua
- Biotechnology and Applied Microbiology Team, Department of Biology, Faculty of Sciences, Abdelmalek-Essaadi University, Tetouan, Morocco
| | - Sara Aboulaghras
- Physiology and Physiopathology Team, Genomic of Human Pathologies Research, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Alia Bouhda
- Department of Medical Oncology, Faculty of Medicine and Pharmacy, Ahmad Bin Zayed Al Nahyan Center of Cancer Treatment, Mohammed VI University Hospital of Tangier, Abdelmalek Essaadi University, Tangier, Morocco
| | - Fatima Zahra El Mrabet
- Department of Medical Oncology, Faculty of Medicine and Pharmacy, Ahmad Bin Zayed Al Nahyan Center of Cancer Treatment, Mohammed VI University Hospital of Tangier, Abdelmalek Essaadi University, Tangier, Morocco
| |
Collapse
|
602
|
Yu H, Zhu W, Lin C, Jia M, Tan X, Yuan Z, Feng S, Yan P. Stromal and tumor immune microenvironment reprogramming through multifunctional cisplatin-based liposomes boosts the efficacy of anti-PD-1 immunotherapy in pancreatic cancer. Biomater Sci 2023; 12:116-133. [PMID: 37921708 DOI: 10.1039/d3bm01118f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
The dense stromal barrier in pancreatic cancer tissues blocks intratumoral delivery and distribution of chemotherapeutics and therapeutic antibodies, causing poor chemoimmunotherapy responses. We designed a multi-targeted pH-sensitive liposome which encapsulates cisplatin (Pt) in its water core (denoted as ATF@Pt Lps) and shows high affinity for uPAR receptors in pancreatic cancer cells, tumor-associated macrophages, and cancer-associated fibroblasts. Systemic administration of ATF@Pt Lps enabled overcoming the central stromal cellular barrier and effective drug delivery into tumor cells, resulting in a strong therapeutic response in a Panc02 cell derived transplanted tumor mouse model. More importantly, ATF@Pt Lps degradation of collagen contributes to the infiltration of CD8+ T cells into tumors as well as an enhanced accumulation of anti PD-1 monoclonal antibodies. Furthermore, the killing of tumor cells by Pt also leads to the release of tumor antigens, which promote the proliferation of immune cells, especially CD83+ cells, Th1 CD4+ cells, and CD8+ cytotoxic T cells, that converted an immunoscore "cold" pancreatic cancer into a pro-immune "hot" tumor. A further combination with an immune checkpoint agent, anti PD-1 antibodies that inhibit PD-1, can enhance tumor specific cytotoxic T cell response. Accordingly, ATF@Pt Lps displays multi-targeting, controlled drug release, stromal disruption, enhanced penetration, killing of cancer cells, modification of the immunosuppressive microenvironment, and enhancement of immunity. This study provides important mechanistic information for the further development of a combination of ATF@Pt Lps and anti PD-1 antibodies for the effective treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Hang Yu
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Wenting Zhu
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Caiyan Lin
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Menglei Jia
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Xiaoxiao Tan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Zhongwen Yuan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Senglin Feng
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| | - Pengke Yan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangzhou, 510150, China.
| |
Collapse
|
603
|
Stosic K, Senar OA, Tarfouss J, Bouchart C, Navez J, Van Laethem JL, Arsenijevic T. A Comprehensive Review of the Potential Role of Liquid Biopsy as a Diagnostic, Prognostic, and Predictive Biomarker in Pancreatic Ductal Adenocarcinoma. Cells 2023; 13:3. [PMID: 38201207 PMCID: PMC10778087 DOI: 10.3390/cells13010003] [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: 10/30/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma is one of the most lethal malignant diseases, with a mortality rate being close to incidence. Due to its heterogeneity and plasticity, as well as the lack of distinct symptoms in the early phases, it is very often diagnosed at an advanced stage, resulting in poor prognosis. Traditional tissue biopsies remain the gold standard for making a diagnosis, but have an obvious disadvantage in their inapplicability for frequent sampling. Blood-based biopsies represent a non-invasive method which potentially offers easy and repeated sampling, leading to the early detection and real-time monitoring of the disease and hopefully an accurate prognosis. Given the urgent need for a reliable biomarker that can estimate a patient's condition and response to an assigned treatment, blood-based biopsies are emerging as a potential new tool for improving patients' survival and surveillance. In this article, we discuss the current advances and challenges in using liquid biopsies for pancreatic cancer, focusing on circulating tumour DNA (ctDNA), extracellular vesicles (EVs), and circulating tumour cells (CTCs), and compare the performance and reliability of different biomarkers and combinations of biomarkers.
Collapse
Affiliation(s)
- Kosta Stosic
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
| | - Oier Azurmendi Senar
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
| | - Jawad Tarfouss
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
| | - Christelle Bouchart
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
- Department of Radiation Oncology, Hopital Universitaire de Bruxelles (H.U.B.), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Julie Navez
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
- Department of Hepato-Biliary-Pancreatic Surgery, Hopital Universitaire de Bruxelles (H.U.B.), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Jean-Luc Van Laethem
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
- Department of Gastroenterology, Hepatology and Digestive Oncology, Hopital Universitaire de Bruxelles (H.U.B.), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium
| | - Tatjana Arsenijevic
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, 1070 Brussels, Belgium (O.A.S.); (C.B.)
- Department of Gastroenterology, Hepatology and Digestive Oncology, Hopital Universitaire de Bruxelles (H.U.B.), Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium
| |
Collapse
|
604
|
Wahler IL, Damanakis A, Große Hokamp N, Bruns C, Schmidt T. Therapy of Locally Advanced and Oligometastatic Pancreatic Adenocarcinoma. Cancers (Basel) 2023; 15:5881. [PMID: 38136425 PMCID: PMC10741431 DOI: 10.3390/cancers15245881] [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: 11/07/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Pancreatic adenocarcinoma is a lethal disease, and surgical resection remains the only curative treatment option. Unfortunately, upon primary diagnosis, only 15-20% of all patients with pancreatic ductal adenocarcinoma (PDAC) have localized disease that is eligible for operation. The remainder of patients either have borderline resectable or locally advanced disease or present with distant metastasis. In this review, we present a comprehensive overview regarding the current strategies and future directions in the multimodal therapy of locally advanced and oligometastasized pancreatic adenocarcinoma and discuss the benefit of surgery following neoadjuvant therapy in these patients.
Collapse
Affiliation(s)
- Isabell Luisa Wahler
- Department of General, Visceral, Cancer and Transplant Surgery, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (I.L.W.); (A.D.)
| | - Alexander Damanakis
- Department of General, Visceral, Cancer and Transplant Surgery, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (I.L.W.); (A.D.)
| | - Nils Große Hokamp
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital of Cologne, 50923 Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral, Cancer and Transplant Surgery, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (I.L.W.); (A.D.)
| | - Thomas Schmidt
- Department of General, Visceral, Cancer and Transplant Surgery, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (I.L.W.); (A.D.)
| |
Collapse
|
605
|
Huffman BM, Feng H, Parmar K, Wang J, Kapner KS, Kochupurakkal B, Martignetti DB, Sadatrezaei G, Abrams TA, Biller LH, Giannakis M, Ng K, Patel AK, Perez KJ, Singh H, Rubinson DA, Schlechter BL, Andrews E, Hannigan AM, Dunwell S, Getchell Z, Raghavan S, Wolpin BM, Fortier C, D’Andrea AD, Aguirre AJ, Shapiro GI, Cleary JM. A Phase I Expansion Cohort Study Evaluating the Safety and Efficacy of the CHK1 Inhibitor LY2880070 with Low-dose Gemcitabine in Patients with Metastatic Pancreatic Adenocarcinoma. Clin Cancer Res 2023; 29:5047-5056. [PMID: 37819936 PMCID: PMC10842136 DOI: 10.1158/1078-0432.ccr-23-2005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/29/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
PURPOSE Combining gemcitabine with CHK1 inhibition has shown promise in preclinical models of pancreatic ductal adenocarcinoma (PDAC). Here, we report the findings from a phase I expansion cohort study (NCT02632448) investigating low-dose gemcitabine combined with the CHK1 inhibitor LY2880070 in patients with previously treated advanced PDAC. PATIENTS AND METHODS Patients with metastatic PDAC were treated with gemcitabine intravenously at 100 mg/m2 on days 1, 8, and 15, and LY2880070 50 mg orally twice daily on days 2-6, 9-13, and 16-20 of each 21-day cycle. Pretreatment tumor biopsies were obtained from each patient for correlative studies and generation of organoid cultures for drug sensitivity testing and biomarker analyses. RESULTS Eleven patients with PDAC were enrolled in the expansion cohort between August 27, 2020 and July 30, 2021. Four patients (36%) experienced drug-related grade 3 adverse events. No objective radiologic responses were observed, and all patients discontinued the trial by 3.2 months. In contrast to the lack of efficacy observed in patients, organoid cultures derived from biopsies procured from two patients demonstrated strong sensitivity to the gemcitabine/LY2880070 combination and showed treatment-induced upregulation of replication stress and DNA damage biomarkers, including pKAP1, pRPA32, and γH2AX, as well as induction of replication fork instability. CONCLUSIONS No evidence of clinical activity was observed for combined low-dose gemcitabine and LY2880070 in this treatment-refractory PDAC cohort. However, the gemcitabine/LY2880070 combination showed in vitro efficacy, suggesting that drug sensitivity for this combination in organoid cultures may not predict clinical benefit in patients.
Collapse
Affiliation(s)
- Brandon M. Huffman
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Hanrong Feng
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Kalindi Parmar
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Junning Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Kevin S. Kapner
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Bose Kochupurakkal
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - David B. Martignetti
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Golbahar Sadatrezaei
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Thomas A. Abrams
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Leah H. Biller
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Anuj K. Patel
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Kimberly J. Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Harshabad Singh
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Douglas A. Rubinson
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Benjamin L. Schlechter
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Elizabeth Andrews
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Alison M. Hannigan
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Stanley Dunwell
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Zoe Getchell
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Srivatsan Raghavan
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | | | - Alan D. D’Andrea
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Andrew J. Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Geoffrey I. Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - James M. Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
606
|
Del Chiaro M, Sugawara T, Karam SD, Messersmith WA. Advances in the management of pancreatic cancer. BMJ 2023; 383:e073995. [PMID: 38164628 DOI: 10.1136/bmj-2022-073995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Pancreatic cancer remains among the malignancies with the worst outcomes. Survival has been improving, but at a slower rate than other cancers. Multimodal treatment, including chemotherapy, surgical resection, and radiotherapy, has been under investigation for many years. Because of the anatomical characteristics of the pancreas, more emphasis on treatment selection has been placed on local extension into major vessels. Recently, the development of more effective treatment regimens has opened up new treatment strategies, but urgent research questions have also become apparent. This review outlines the current management of pancreatic cancer, and the recent advances in its treatment. The review discusses future treatment pathways aimed at integrating novel findings of translational and clinical research.
Collapse
Affiliation(s)
- Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Toshitaka Sugawara
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sana D Karam
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Wells A Messersmith
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
607
|
Behrens D, Pfohl U, Conrad T, Becker M, Brzezicha B, Büttner B, Wagner S, Hallas C, Lawlor R, Khazak V, Linnebacher M, Wartmann T, Fichtner I, Hoffmann J, Dahlmann M, Walther W. Establishment and Thorough Characterization of Xenograft (PDX) Models Derived from Patients with Pancreatic Cancer for Molecular Analyses and Chemosensitivity Testing. Cancers (Basel) 2023; 15:5753. [PMID: 38136299 PMCID: PMC10741928 DOI: 10.3390/cancers15245753] [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: 09/29/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Patient-derived xenograft (PDX) tumor models are essential for identifying new biomarkers, signaling pathways and novel targets, to better define key factors of therapy response and resistance mechanisms. Therefore, this study aimed at establishing pancreas carcinoma (PC) PDX models with thorough molecular characterization, and the identification of signatures defining responsiveness toward drug treatment. In total, 45 PC-PDXs were generated from 120 patient tumor specimens and the identity of PDX and corresponding patient tumors was validated. The majority of engrafted PDX models represent ductal adenocarcinomas (PDAC). The PDX growth characteristics were assessed, with great variations in doubling times (4 to 32 days). The mutational analyses revealed an individual mutational profile of the PDXs, predominantly showing alterations in the genes encoding KRAS, TP53, FAT1, KMT2D, MUC4, RNF213, ATR, MUC16, GNAS, RANBP2 and CDKN2A. Sensitivity of PDX toward standard of care (SoC) drugs gemcitabine, 5-fluorouracil, oxaliplatin and abraxane, and combinations thereof, revealed PDX models with sensitivity and resistance toward these treatments. We performed correlation analyses of drug sensitivity of these PDX models and their molecular profile to identify signatures for response and resistance. This study strongly supports the importance and value of PDX models for improvement in therapies of PC.
Collapse
Affiliation(s)
- Diana Behrens
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Ulrike Pfohl
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Theresia Conrad
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Michael Becker
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Bernadette Brzezicha
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Britta Büttner
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Silvia Wagner
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Cora Hallas
- Institut für Hämatopathologie, Fangdieckstr. 75, 22547 Hamburg, Germany
| | - Rita Lawlor
- ARC-Net Research Center, University and Hospital Trust of Verona, Piazzale A. Scuro 10, 37134 Verona, Italy
| | | | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, University Medical Center Rostock, 18057 Rostock, Germany
| | - Thomas Wartmann
- University Clinic for General, Visceral, Vascular and Transplantation Surgery, Faculty of Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Iduna Fichtner
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Jens Hoffmann
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Mathias Dahlmann
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Wolfgang Walther
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin, Lindenberger Weg 80, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| |
Collapse
|
608
|
Rogers S, Charles A, Thomas RM. The Prospect of Harnessing the Microbiome to Improve Immunotherapeutic Response in Pancreatic Cancer. Cancers (Basel) 2023; 15:5708. [PMID: 38136254 PMCID: PMC10741649 DOI: 10.3390/cancers15245708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/24/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Pancreatic ductal adenocarcinoma cancer (PDAC) is projected to become the second leading cause of cancer-related death in the United States by 2030. Patients are often diagnosed with advanced disease, which explains the dismal 5-year median overall survival rate of ~12%. Immunotherapy has been successful in improving outcomes in the past decade for a variety of malignancies, including gastrointestinal cancers. However, PDAC is historically an immunologically "cold" tumor, one with an immunosuppressive environment and with restricted entry of immune cells that have limited the success of immunotherapy in these tumors. The microbiome, the intricate community of microorganisms present on and within humans, has been shown to contribute to many cancers, including PDAC. Recently, its role in tumor immunology and response to immunotherapy has generated much interest. Herein, the current state of the interaction of the microbiome and immunotherapy in PDAC is discussed with a focus on needed areas of study in order to harness the immune system to combat pancreatic cancer.
Collapse
Affiliation(s)
- Sherise Rogers
- Department of Medicine, Division of Hematology and Oncology, University of Florida College of Medicine, Gainesville, FL 32610, USA;
| | - Angel Charles
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA;
| | - Ryan M. Thomas
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA;
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32603, USA
| |
Collapse
|
609
|
Surana R, Gonzalez GN, Rogers J, Hong DS, Yap TA, Rodon J, Naing A, Wolff RA, Smaglo BG, Bernstam FM, Subbiah V, Pant S. Utility of Established Prognostic Scoring Systems for Patients with Advanced Pancreatic Adenocarcinoma Enrolled in Immunotherapy-Based Early-Phase Clinical Trials. J Gastrointest Cancer 2023; 54:1308-1315. [PMID: 37119430 DOI: 10.1007/s12029-023-00930-7] [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] [Accepted: 03/19/2023] [Indexed: 05/01/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy for which multiagent chemotherapy is the mainstay of treatment resulting in limited survival and symptomatic benefit. Treatment with immune checkpoint inhibitors (ICI) has proven effective in a growing number of solid tumors but has yet to show clinical benefit in patients with PDAC. Given the growing number of ICI-based clinical trials in development for patients with PDAC and lack of clinical benefit thus far with ICI-based therapies in these patients, we sought to (1) determine the outcomes of patients with PDAC treated with ICI-based therapies as part of an early phase clinical trial, (2) validate the utility of established prognostic scoring systems, and (3) identify novel prognostic factors in an attempt to better identify patients that would benefit from enrollment onto an ICI-based early phase clinical trial. METHODS We conducted a single-center retrospective analysis of patients with advanced PDAC who were treated with ICI-based therapy as part of an early-phase clinical trial. RESULTS Patients were only able to stay on study for a limited time due to disease progression and/or a change in performance status and had a poor overall survival. Established prognostic scoring systems were not effective in predicting outcomes in this patient population, but factors such as pre-treatment albumin neutrophil to lymphocyte ratio (NLC) may be helpful in patient selection. CONCLUSIONS This study underscores the need for larger studies to help identify patient and tumor intrinsic factors that predict response to ICI-based therapies in patients with PDAC.
Collapse
Affiliation(s)
- Rishi Surana
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Jane Rogers
- Pharmacy Clinical Programs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brandon G Smaglo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shubham Pant
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
610
|
Dallmann J, Freitag J, Jung C, Khinvasara K, Merz L, Peters D, Schork M, Beck J. CIMT 2023: report on the 20th Annual Meeting of the Association for Cancer Immunotherapy. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 20:100397. [PMID: 37876518 PMCID: PMC10590812 DOI: 10.1016/j.iotech.2023.100397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
The Association for Cancer Immunotherapy (CIMT) celebrated the 20th anniversary of the CIMT Annual Meeting. CIMT2023 was held 3-5 May 2023 in Mainz, Germany. 1051 academic and clinical professionals from over 30 countries attended the meeting and discussed the latest advances in cancer immunology and immunotherapy research. This report summarizes the highlights of CIMT2023.
Collapse
Affiliation(s)
- J. Dallmann
- Immunotherapies & Preclinical Research, BioNTech SE, Mainz
| | - J. Freitag
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - C. Jung
- BioNTech Cell & Gene Therapies GmbH, Mainz
| | - K. Khinvasara
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - L. Merz
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - D. Peters
- Immunotherapies & Preclinical Research, BioNTech SE, Mainz
| | - M. Schork
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - J.D. Beck
- Immunotherapies & Preclinical Research, BioNTech SE, Mainz
| |
Collapse
|
611
|
Augustinus S, Broekman T, Creemers GJ, Daamen LA, van Dieren S, de Groot JWB, Cirkel GA, Homs MYV, van Laarhoven HWM, van Leeuwen L, Los M, Luelmo SAC, van Oijen MGH, Spierings LEAM, de Vos-Geelen J, Besselink MG, Wilmink JW. Timing of start of systemic treatment in patients with asymptomatic metastasized pancreatic cancer (TIMEPAN): a protocol of a multicenter prospective patient preference non-randomized trial. Acta Oncol 2023; 62:1973-1978. [PMID: 37897803 DOI: 10.1080/0284186x.2023.2273898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/17/2023] [Indexed: 10/30/2023]
Affiliation(s)
- Simone Augustinus
- Amsterdam UMC, Department of Surgery, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Thijmen Broekman
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, Department of Medical Oncology, University of Amsterdam, Amsterdam, The Netherlands
| | - Geert-Jan Creemers
- Department of Medical Oncology, Catherina Hospital, Eindhoven, The Netherlands
| | - Lois A Daamen
- Division of Imaging & Oncology, University Medical Center Utrecht Cancer Center, Utrecht University, Utrecht, The Netherlands
- Department of Surgery, University Medical Center Utrecht Cancer Center, St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Susan van Dieren
- Amsterdam UMC, Department of Surgery, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Geert A Cirkel
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, Meander Medisch Centrum, Amersfoort, The Netherlands
| | - Marjolein Y V Homs
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hanneke W M van Laarhoven
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, Department of Medical Oncology, University of Amsterdam, Amsterdam, The Netherlands
| | - Lobke van Leeuwen
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, Diakonessenhuis, Utrecht, The Netherlands
| | - Maartje Los
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, St. Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Saskia A C Luelmo
- Department of Medical Oncology, Leids Universitair Medisch Centrum, Leiden, The Netherlands
| | - Martijn G H van Oijen
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, Department of Medical Oncology, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Judith de Vos-Geelen
- Department of Medical Oncology, GROW, Maastricht UMC, Maastsricht, The Netherlands
| | - Marc G Besselink
- Amsterdam UMC, Department of Surgery, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Johanna W Wilmink
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, Department of Medical Oncology, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
612
|
Grierson PM, Suresh R, Tan B, Pedersen KS, Amin M, Park H, Trikalinos NA, Liu J, Boice N, Brown A, Bansod S, Wang-Gillam A, Lim KH. A Pilot Study of Paricalcitol plus Nanoliposomal Irinotecan and 5-FU/LV in Advanced Pancreatic Cancer Patients after Progression on Gemcitabine-Based Therapy. Clin Cancer Res 2023; 29:4733-4739. [PMID: 37801295 PMCID: PMC11123290 DOI: 10.1158/1078-0432.ccr-23-1405] [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: 05/13/2023] [Revised: 07/27/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
PURPOSE Vitamin D analogues remodel the desmoplastic stroma, and improve vascularity and efficacy of chemotherapy in preclinical pancreas cancer models. PATIENTS AND METHODS We conducted a pilot study to evaluate the safety and preliminary efficacy of the vitamin D analogue paricalcitol in combination with nanoliposomal irinotecan (Nal-iri) plus 5-fluorouracil/leucovorin (5-FU/LV) in patients with advanced pancreatic cancer who had progressed on gemcitabine-based therapy. Two dose levels (DL) of paricalcitol were tested: fixed dose weekly (75 mcg, DL1) and weight-based weekly (7 mcg/kg, /DL2). The primary endpoint was safety, and secondary endpoints included overall response rate, progression-free survival (PFS), and overall survival (OS). Correlative objectives aimed to identify molecular predictors of response and alterations in the tumor stroma. RESULTS Twenty patients (10 each in DL1 and DL2) enrolled between March 2019 and May 2021. No grade 3/4 adverse events related to paricalcitol were observed. The most common toxicities were nausea, diarrhea and fatigue, which were similar in both cohorts. Three patients discontinued study after one cycle and were not radiographically evaluable. Of the remaining 17 evaluable patients, 2 had partial response and 12 had stable disease. The median PFS for response-evaluable patients in DL1 was 4.14 months, for DL2 was 4.83 months. Intent-to-treat median OS was 6.15 and 6.66 months for DL1 and DL2, respectively. Correlative studies showed increased tumor vascularity in posttreatment samples in patients receiving the higher dose of paricalcitol (DL2). CONCLUSIONS Paricalcitol at 7 mcg/kg/week in combination with Nal-iri/ 5-FU/LV is safely tolerated, may increase tumor vascularity and warrants further investigation.
Collapse
Affiliation(s)
- Patrick M. Grierson
- Division of Medical Oncology, Department of Internal Medicine, Washington University, St. Louis, MO, USA
| | - Rama Suresh
- Division of Medical Oncology, Department of Internal Medicine, Washington University, St. Louis, MO, USA
| | - Benjamin Tan
- Division of Medical Oncology, Department of Internal Medicine, Washington University, St. Louis, MO, USA
| | - Katrina S. Pedersen
- Division of Medical Oncology, Department of Internal Medicine, Washington University, St. Louis, MO, USA
| | - Manik Amin
- Section of Hematology/Oncology, Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, NH
| | - Haeseong Park
- Division of Medical Oncology, Department of Internal Medicine, Washington University, St. Louis, MO, USA
| | - Nikolaos A Trikalinos
- Division of Medical Oncology, Department of Internal Medicine, Washington University, St. Louis, MO, USA
| | - Jingxia Liu
- Division of Public Health Sciences, Department of Surgery Washington University, St. Louis, MO
| | - Nicholas Boice
- Division of Medical Oncology, Department of Internal Medicine, Washington University, St. Louis, MO, USA
| | - Amberly Brown
- Division of Medical Oncology, Department of Internal Medicine, Washington University, St. Louis, MO, USA
| | - Sapana Bansod
- Division of Medical Oncology, Department of Internal Medicine, Washington University, St. Louis, MO, USA
| | - Andrea Wang-Gillam
- Division of Medical Oncology, Department of Internal Medicine, Washington University, St. Louis, MO, USA
| | - Kian-Huat Lim
- Division of Medical Oncology, Department of Internal Medicine, Washington University, St. Louis, MO, USA
| |
Collapse
|
613
|
Nicolais LM, Mohamed A, Macgillivray D, Verdini N, Inhorn R, Dugan M, Hayward CM, Fitzgerald TL. Improved Overall Survival of Patients with Pancreatic Cancer through Multiagent Chemotherapy and Increased Rates of Surgical Resection. Am Surg 2023; 89:5964-5971. [PMID: 37295019 DOI: 10.1177/00031348221148350] [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] [Indexed: 06/11/2023]
Abstract
BACKGROUND Seminal trials have demonstrated improved survival in pancreatic adenocarcinoma with novel multiagent chemotherapy regimens. To understand the clinical ramifications of this paradigm shift, we reviewed our institutional experience. METHODS This retrospective cohort study utilized a prospective database at a single institution to study all patients diagnosed with and treated for pancreatic adenocarcinoma between 2000 and 2020. RESULTS 1,572 patients were included of which 36% were diagnosed before (Era 1) and 64% after (Era 2) 2011. Survival improved in Era 2 (Median survival 10 vs 8 months, HR .79; P < .001). The survival advantage for Era 2 was primarily seen in patients with high-risk disease (12 vs10 months, HR .71; P < .001). A similar trend was noted for patients undergoing surgical resection (26 vs 21 months, HR .80; P = .081) and with imminently resectable tumors (19 vs 15 months, HR .88; P = .4); however, this was not statistically significant. There was no survival advantage for patients with stage IV disease (4 vs 4 months). Patients in Era 2 were more likely to undergo surgery (OR 2.78; CI 2.00-3.92, P < .001). This increase was driven primarily by increased surgical resection for those with high-risk disease (42 vs 20%, OR 3.74; P < .001). DISCUSSION/CONCLUSIONS This single institutional study showed improved survival after the shift to novel chemotherapy regimens. This was driven by improved survival for patients with high-risk disease and may be due to more effective eradication of microscopic metastatic disease with adjuvant chemotherapy and increased resection rates.
Collapse
Affiliation(s)
- Laura M Nicolais
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
- Graduate School of Biomedical Sciences, Tufts University Clinical and Translational Science Graduate Program, Boston, MA, USA
| | | | - Dougald Macgillivray
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
| | - Nicholas Verdini
- Tufts University School of Medicine, Washington St, Boston, MA, USA
- Boston Medical Center, Boston, MA, USA
| | - Roger Inhorn
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
| | - Matthew Dugan
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
| | - Cynthia M Hayward
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
| | - Timothy L Fitzgerald
- Division of Surgical Oncology, Tufts University School of Medicine Maine Medical Center, Portland, ME, USA
| |
Collapse
|
614
|
Al Abbas AI, Meier J, Hester CA, Radi I, Yan J, Zhu H, Mansour JC, Porembka MR, Wang SC, Yopp AC, Zeh HJ, Polanco PM. Impact of Neoadjuvant Treatment and Minimally Invasive Surgery on Perioperative Outcomes of Pancreatoduodenectomy: an ACS NSQIP Analysis. J Gastrointest Surg 2023; 27:2823-2842. [PMID: 37903972 DOI: 10.1007/s11605-023-05859-7] [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: 04/24/2023] [Accepted: 08/31/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND There is an increasing use of neoadjuvant treatment (NAT) for pancreatic cancer (PC) followed by minimally invasive pancreatoduodenectomy (MIPD). We evaluate the impact of the surgical approach on 30-day outcomes in PC patients who underwent NAT. METHODS Patients with PC who had NAT followed by MIPD or open pancreatoduodenectomy (OPD) were identified from a pancreatectomy-targeted dataset (2014-2020) of the National Surgical Quality Improvement Program. Comparisons were made between MIPD and OPD within NAT groups. RESULTS A total of 5588 patients were analyzed. Of those, 4907 underwent OPD and 476 underwent MIPD. In addition, 3559 patients received neoadjuvant chemotherapy alone and 1830 received neoadjuvant chemoradiation. In the chemotherapy-alone group, the MIPD subgroup had lower rates of any complication (38.2% vs. 45.8%, P = 0.005), but there were no differences in mortality (2.1% for MIPD vs 1.9% for OPD, P=0.8) or serious complication (11.8% for MIPD vs 15% for OPD, P=0.1). On multivariable analysis, MIPD was independently predictive of lower rates of any complication (OR: 0.74, 95% CI 0.6-0.93, P = 0.0009), CR-POPF (OR: 0.58, 95% CI 0.35-0.96, P = 0.04), and shorter LOS (estimate: -1.03, 95% CI -1.73 to -0.32, P = 0.004). In the chemoradiation group, patients undergoing MIPD had higher rates of preoperative diabetes (P < 0.05), but there were no significant differences in any outcomes between the two approaches in this group. CONCLUSION MIPD is safe and feasible after NAT. Patients having neoadjuvant chemotherapy alone followed by MIPD had lower rates of complications, shorter LOS, and fewer CR-POPFs compared to OPD.
Collapse
Affiliation(s)
- Amr I Al Abbas
- University of Texas Southwestern, Department of Surgery, Dallas, TX, USA
| | - Jennie Meier
- University of Texas Southwestern, Department of Surgery, Dallas, TX, USA
| | - Caitlin A Hester
- University of Texas Southwestern, Department of Surgery, Dallas, TX, USA
| | - Imad Radi
- University of Texas Southwestern, Harold C. Simmons Cancer Center, Dallas, TX, USA
| | - Jinsheng Yan
- University of Texas Southwestern, Harold C. Simmons Cancer Center, Dallas, TX, USA
| | - Hong Zhu
- University of Texas Southwestern, Harold C. Simmons Cancer Center, Dallas, TX, USA
| | - John C Mansour
- University of Texas Southwestern, Harold C. Simmons Cancer Center, Dallas, TX, USA
| | - Matthew R Porembka
- University of Texas Southwestern, Harold C. Simmons Cancer Center, Dallas, TX, USA
| | - Sam C Wang
- University of Texas Southwestern, Harold C. Simmons Cancer Center, Dallas, TX, USA
| | - Adam C Yopp
- University of Texas Southwestern, Harold C. Simmons Cancer Center, Dallas, TX, USA
| | - Herbert J Zeh
- University of Texas Southwestern, Department of Surgery, Dallas, TX, USA
- University of Texas Southwestern, Harold C. Simmons Cancer Center, Dallas, TX, USA
| | - Patricio M Polanco
- University of Texas Southwestern, Department of Surgery, Dallas, TX, USA.
- University of Texas Southwestern, Harold C. Simmons Cancer Center, Dallas, TX, USA.
- Division of Surgical Oncology, University of Texas Southwestern Medical Center, 2201 Inwood Road, 3rd Floor, Dallas, TX, 75390, USA.
| |
Collapse
|
615
|
D'Angelo A, Catalano M, Conca R, Petrioli R, Siminonato F, Cappetta A, Roviello G, Ramello M. Clinical outcome and safety profile of metastatic pancreatic cancer patients treated with more than six cycles of nab-paclitaxel plus gemcitabine. J Chemother 2023; 35:753-759. [PMID: 37167100 DOI: 10.1080/1120009x.2023.2190712] [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: 06/13/2022] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 05/13/2023]
Abstract
The phase III MPACT trial demonstrated the superiority of gemcitabine plus nab-paclitaxel (NABGEM) versus gemcitabine alone in previously untreated patients with metastatic pancreatic cancer (mPC). The aim of this study was to evaluate the responses in terms of efficacy and safety in patients treated with more than 6 cycles of chemotherapy. From January 2015 to December 2018, patients with mPC receiving first-line treatment with NABGEM were included in a multicentre retrospective observational study. Exploratory analyses of efficacy and safety were performed. The cohort included 153 patients with performance status of 1. The median overall survival and progression-free survival were 20 months (hazard ratio [HR] 0.28, 95% confidence interval [CI]: 0.17-0.44) and 10 months (HR 0.24 95% CI: 0.16-0.38) respectively, in patients who received >6 cycles compared to 9 and 5 months in those treated with ≤6 cycles (p < 0.001). The disease control rate was 100% versus 56% in patients receiving >6 and ≤6 cycles, respectively. No progression of disease was recorded in patients who received >6 cycles. Grade 1 neuropathy and grade 3 neutropenia were more frequent in patients treated with >6 cycles compared to patients receiving ≤6 cycles (p = 0.01; p = 0.03, respectively). Dose reduction was necessary for 70.1% and 53.4% of patients treated with >6 or ≤6 cycles, whereas treatment interruption occurred in 37.1% and 21.6%, respectively. Our results confirmed the efficacy and safety of NABGEM in untreated mPC. In particular, we highlighted significant clinical efficacy in patients who received >6 cycles of chemotherapy compared to those who received ≤6 cycles, with manageable toxicity profile.
Collapse
Affiliation(s)
- Alberto D'Angelo
- Department of Life Sciences, University of Bath, Bath, UK
- Department of Oncology, Royal United Hospital, Bath, UK
| | - Martina Catalano
- School of Human Health Sciences, University of Florence, Florence, Italy
| | - Raffaele Conca
- Division of Medical Oncology, Department of Onco-Hematology, IRCCS-CROB, Referral Cancer Center of Basilicata, Vulture, Potenza, Italy
- Department of Oncology, San Bortolo General Hospital, Vicenza, Italy
| | - Roberto Petrioli
- Department of Medicine, Surgery and Neurosciences, Medical Oncology Unit, University of Siena, Siena, Italy
| | | | | | | | - Monica Ramello
- Oncology Unit, Department of Medical, Surgical, & Health Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
616
|
Miyata Y, Yonamine N, Fujinuma I, Tsunenari T, Takihata Y, Hakoda H, Nakazawa A, Iwasaki T, Einama T, Togashi J, Tsujimoto H, Ueno H, Beck Y, Kishi Y. Impact of Preoperative Tumor Size on Prognosis of Resectable and Borderline Resectable Pancreatic Ductal Adenocarcinomas. Ann Surg Oncol 2023; 30:8621-8630. [PMID: 37658273 DOI: 10.1245/s10434-023-14219-3] [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: 11/25/2022] [Accepted: 08/08/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND Tumor size (TS) is a well-established prognostic factor of pancreatic ductal adenocarcinoma (PDAC). However, whether a uniform treatment strategy can be applied for all resectable PDACs (R-PDACs) and borderline resectable PDACs (BR-PDACs), regardless of TS, remains unclear. This study aimed to investigate the impact of preoperative TS on surgical outcomes of patients with R-PDACs and BR-PDACs. METHODS Chart data from three institutions were reviewed to select patients who underwent pancreatectomy for R-PDACs and BR-PDACs between January 2006 and December 2020. The patients were divided into TSsmall and TSlarge groups according to a TS cutoff value determined for each of R- and BR-PDAC using the minimum P value approach for the risk of R1 resection. RESULTS TS of 35 mm and 24 mm was the best cutoff value in R-PDAC and BR-PDAC, respectively. The R1 rate was higher in the TSlarge than TSsmall group, in both R- (n = 35, 37% versus n = 294, 19%; P = 0.011) and BR-PDAC (n = 89, 37% versus n = 27, 15%; P = 0.030). Overall survival was significantly better in the TSsmall than TSlarge group in R-PDAC (38.2 versus 12.1 months; P < 0.001), but comparable between the two groups in BR-DPAC (21.2 versus 22.7 months; P = 0.363). Multivariate analysis revealed TS > 35 mm as an independent predictor of worse survival in patients with R-PDAC. CONCLUSION Larger TS was associated with a higher R1 rate and is a worse prognostic factor in patients with R-PDAC.
Collapse
Affiliation(s)
- Yoichi Miyata
- Department of Surgery, Asahi General Hospital, Asahi, Chiba, Japan
| | - Naoto Yonamine
- Department of Surgery, National Defense Medical College Hospital, Tokorozawa, Saitama, Japan
| | - Ibuki Fujinuma
- Department of Surgery, National Defense Medical College Hospital, Tokorozawa, Saitama, Japan
| | - Takazumi Tsunenari
- Department of Surgery, National Defense Medical College Hospital, Tokorozawa, Saitama, Japan
| | - Yasuhiro Takihata
- Department of Surgery, National Defense Medical College Hospital, Tokorozawa, Saitama, Japan
| | - Hiroyuki Hakoda
- Department of Surgery, Asahi General Hospital, Asahi, Chiba, Japan
| | - Akiko Nakazawa
- Department of Surgery, National Defense Medical College Hospital, Tokorozawa, Saitama, Japan
| | - Toshimitsu Iwasaki
- Department of Surgery, National Defense Medical College Hospital, Tokorozawa, Saitama, Japan
| | - Takahiro Einama
- Department of Surgery, National Defense Medical College Hospital, Tokorozawa, Saitama, Japan
| | - Junichi Togashi
- Department of Surgery, Asahi General Hospital, Asahi, Chiba, Japan
| | - Hironori Tsujimoto
- Department of Surgery, National Defense Medical College Hospital, Tokorozawa, Saitama, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College Hospital, Tokorozawa, Saitama, Japan
| | - Yoshifumi Beck
- Department of Hepatobiliary pancreatic surgery, Saitama Medical Center, Saitama, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College Hospital, Tokorozawa, Saitama, Japan.
| |
Collapse
|
617
|
Kitamura H, Morizane C, Tanabe N, Go I, Maruki Y, Ohba A, Nagashio Y, Kondo S, Hijioka S, Ueno H, Yoshida T, Okusaka T. Clinical features of germline BRCA1/2 or ATM pathogenic variant positive pancreatic cancer in Japan. Pancreatology 2023; 23:964-969. [PMID: 37914629 DOI: 10.1016/j.pan.2023.10.017] [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: 07/03/2023] [Revised: 10/08/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND There has been increasing interest into the role of germline BRCA1/2 pathogenic variants (gBRCA PV) and gATM PV and likely PV (PV and LPV; PV + LPV) in the carcinogenesis and treatment of pancreatic cancer (PC), but the clinical features have not been well described. METHODS Patients with confirmed gBRCA PV and gATM PV + LPV PC treated at our hospital between April 2016 and December 2021, were retrospectively evaluated for clinical characteristics and outcomes. RESULTS Twenty-two patients harbored gBRCA PV and three patients harbored gATM PV + LPV. Of the gBRCA PV patients, 81.8 % received platinum-based chemotherapy with favorable treatment outcomes with an objective response rate of 50.0 % (95 % CI: 23.0-77.0), median progression free survival (PFS) of 334 days, and median overall survival (OS) of 926 days from the initiation of first-line chemotherapy. The annual number of patients with gBRCA PV was two patients per year before January 2021 (when BRACAnalysis became available in Japan), and ten patients during the 10 months thereafter. Four patients (20 %) with gBRCA PV developed soft-tissue metastasis with progression. Two patients with gATM PV + LPV received platinum-based chemotherapy and the best response of those patients was partial response and stable disease and their OS from the initiation of first-line chemotherapy was 1192 and 989 days, and PFS was 579 and 140 days, respectively. CONCLUSION The diagnosis of gBRCA PV-positive PC has increased revealed in recent years. These tumors appear to be sensitive to platinum-based chemotherapy, with long term survival observed in gATM PV + LPV-positive patients.
Collapse
Affiliation(s)
- Hidetoshi Kitamura
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan.
| | - Noriko Tanabe
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Japan
| | - Ikeda Go
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Yuta Maruki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Akihiro Ohba
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Yoshikuni Nagashio
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Shunsuke Kondo
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Susumu Hijioka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Hideki Ueno
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Teruhiko Yoshida
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| |
Collapse
|
618
|
Stupan U, Čemažar M, Trotovšek B, Petrič M, Tomažič A, Gašljević G, Ranković B, Seliškar A, Plavec T, Sredenšek J, Plut J, Štukelj M, Lampreht Tratar U, Jesenko T, Nemec Svete A, Serša G, Đokić M. Histologic changes of porcine portal vein anastomosis after electrochemotherapy with bleomycin. Bioelectrochemistry 2023; 154:108509. [PMID: 37459749 DOI: 10.1016/j.bioelechem.2023.108509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/19/2023] [Accepted: 07/09/2023] [Indexed: 09/16/2023]
Abstract
Electrochemotherapy (ECT1) is used for treatment of unresectable abdominal malignancies. This study aims to show that ECT of porcine portal vein anastomosis is safe and feasible in order to extend the indications for margin attenuation after resection of locally advanced pancreatic carcinoma. No marked differences were found between the control group and ECT treated groups. Electroporation thus caused irreversible damage to the vascular smooth muscle cells in tunica media that could bedue to the narrow irreversible electroporation zone that may occur near the electrodes, or due to vasa vasorum thrombosis in the tunica externa. Based on the absence of vascular complications, and similar histological changes in lienal veinanastomosis, we can conclude that ECT of portal vein anastomosis is safe and feasible.
Collapse
Affiliation(s)
- Urban Stupan
- University of Ljubljana, Faculty of Medicine, Korytkova ulica 2, SI-1000 Ljubljana, Slovenia.
| | - Maja Čemažar
- Institute of Oncology Ljubljana, Zaloška cesta 2, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Veterinary Faculty, Gerbičeva ulica 60, SI-1000 Ljubljana, Slovenia
| | - Blaž Trotovšek
- University of Ljubljana, Faculty of Medicine, Korytkova ulica 2, SI-1000 Ljubljana, Slovenia; University Medical Centre Ljubljana, Zaloška cesta 2, SI-1000 Ljubljana, Slovenia
| | - Miha Petrič
- University of Ljubljana, Faculty of Medicine, Korytkova ulica 2, SI-1000 Ljubljana, Slovenia; University Medical Centre Ljubljana, Zaloška cesta 2, SI-1000 Ljubljana, Slovenia
| | - Aleš Tomažič
- University of Ljubljana, Faculty of Medicine, Korytkova ulica 2, SI-1000 Ljubljana, Slovenia; University Medical Centre Ljubljana, Zaloška cesta 2, SI-1000 Ljubljana, Slovenia
| | - Gorana Gašljević
- Institute of Oncology Ljubljana, Zaloška cesta 2, SI-1000 Ljubljana, Slovenia
| | - Branislava Ranković
- University of Ljubljana, Faculty of Medicine, Korytkova ulica 2, SI-1000 Ljubljana, Slovenia
| | - Alenka Seliškar
- University of Primorska, Faculty of Health Sciences, Polje 42, SI-6310 Izola, Slovenia
| | - Tanja Plavec
- University of Primorska, Faculty of Health Sciences, Polje 42, SI-6310 Izola, Slovenia
| | - Jerneja Sredenšek
- University of Primorska, Faculty of Health Sciences, Polje 42, SI-6310 Izola, Slovenia
| | - Jan Plut
- University of Primorska, Faculty of Health Sciences, Polje 42, SI-6310 Izola, Slovenia
| | - Marina Štukelj
- University of Primorska, Faculty of Health Sciences, Polje 42, SI-6310 Izola, Slovenia
| | | | - Tanja Jesenko
- Institute of Oncology Ljubljana, Zaloška cesta 2, SI-1000 Ljubljana, Slovenia
| | - Alenka Nemec Svete
- University of Primorska, Faculty of Health Sciences, Polje 42, SI-6310 Izola, Slovenia
| | - Gregor Serša
- Institute of Oncology Ljubljana, Zaloška cesta 2, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Health Sciences, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| | - Mihajlo Đokić
- University of Ljubljana, Faculty of Medicine, Korytkova ulica 2, SI-1000 Ljubljana, Slovenia; University Medical Centre Ljubljana, Zaloška cesta 2, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
619
|
Leonhardt CS, Stamm T, Hank T, Prager G, Strobel O. Defining oligometastatic pancreatic cancer: a systematic review and critical synthesis of consensus. ESMO Open 2023; 8:102067. [PMID: 37988953 PMCID: PMC10774968 DOI: 10.1016/j.esmoop.2023.102067] [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: 08/05/2023] [Accepted: 10/14/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Small retrospective series suggest that local consolidative treatment (LCT) may improve survival in oligometastatic pancreatic ductal adenocarcinoma (PDAC). However, no uniform definition of oligometastatic disease (OMD) in PDAC exists; this impedes meaningful conclusions. PATIENTS AND METHODS A systematic literature search using PubMed, Web of Science, and Cochrane CENTRAL registries for studies and protocols reporting on definitions and/or LCT of OMD in PDAC was performed. The primary endpoint was the definition of OMD. Levels of agreement were categorized as consensus (≥75% agreement between studies), fair agreement (50%-74%), and absent/poor agreement (<50%). RESULTS After screening of 5374 abstracts, the full text of 218 studies was assessed, of which 76 were included in the qualitative synthesis. The majority of studies were retrospective (n = 66, 87%), two were prospective studies and eight were study protocols. Studies investigated mostly liver (n = 38, 51%) and lung metastases (n = 15, 20%). Across studies, less than one-half (n = 32, 42%) reported a definition of OMD, while 44 (58%) did not. Involvement was limited to a single organ (consensus). Additional criteria for defining OMD were the number of lesions (consensus), metastatic site (poor agreement), metastatic size (poor agreement), treatment possibilities (poor agreement), and biomarker response (poor agreement). Liver OMD could involve three or fewer lesions (consensus) and synchronous disease (fair agreement), while lung metastases could involve two or fewer lesions and metachronous disease (consensus). The large majority of studies were at a high risk of bias or did not include any control groups. CONCLUSION Definitions of OMD were not used or varied widely between studies hampering across-study comparability and highlighting an unmet need for a consensus. The present study is part of a multistep process that aims to develop an interdisciplinary consensus on OMD in pancreatic cancer.
Collapse
Affiliation(s)
- C-S Leonhardt
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna
| | - T Stamm
- Institute of Outcomes Research, Center for Medical Data Science, Medical University of Vienna; Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna
| | - T Hank
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna
| | - G Prager
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - O Strobel
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna.
| |
Collapse
|
620
|
Halder R, Veeravelli S, Cheng C, Estrada-Mendizabal RJ, Recio-Boiles A. Health Disparities in Presentation, Treatment, Genomic Testing, and Outcomes of Pancreatic Cancer in Hispanic and Non-Hispanic Patients. J Racial Ethn Health Disparities 2023; 10:3131-3139. [PMID: 37071331 PMCID: PMC10645638 DOI: 10.1007/s40615-022-01486-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 11/18/2022] [Accepted: 12/02/2022] [Indexed: 04/19/2023]
Abstract
BACKGROUND There are few conflicting results regarding the treatment and outcomes of Hispanic patients with pancreatic cancer. This study comprehensively evaluated the differences in baseline characteristics, treatments, genomic testing, and outcomes among Hispanic (H) and Non-Hispanic (NH) patients with early-stage (ES) and late-stage (LS) pancreatic cancer (PC). METHODS This is a retrospective analysis from 2013 to 2020 of 294 patients with pancreatic ductal adenocarcinoma; data collected included patient demographics, clinical characteristics, treatment regimens, response, germline and somatic genetic testing, and survival outcomes. Excluded those with insufficient data. Univariate comparisons used parametric and nonparametric tests as appropriate to evaluate for differences between H and NH groups. Fisher's exact tests were performed to evaluate the difference in frequency. Kaplan-Meier and Cox regression analysis assessed the survival. RESULTS The analysis included 198 patients who had a late-stage disease and 96 patients with early-stage disease at the time of diagnosis. Among the early-stage patients, the median age at diagnosis was 60.7 years in the H versus 66.7 years in the NH (p = 0.03). No other differences were observed in baseline characteristics, treatments offered, and median overall survival (NH 25 vs. H 17.7 months, p = 0.28). Performance status, negative surgical margins, and adjuvant therapy were clinically significant and univariable with improved OS (p < 0.05), regardless of ethnicity. Hispanic patients with early pancreatic cancer were noted to be at a greater risk of death with a statistically significant hazard ratio of 3.1 (p = 0.005, 95% CI, 1.39-6.90). Among the late-stage patients, Hispanic patients with ≥ 3 predisposing risk factors for pancreatic cancer were 44% vs. 25% of NH (p = 0.006). No significant differences were noted in baseline characteristic treatments, progression-free, and median overall survivals (NH 10.0 vs. 9.2 months, p = 0.4577). In the late-stage genomic testing, germline testing performed in NH 69.4% vs. H 43.9% (p = 0.003) revealed no difference among groups. For the somatic testing, the pathogenic variants with actionable mutations were 2.5% of NH and 17.6% of H patients (p = 0.03). CONCLUSION Hispanic patients with early-stage pancreatic adenocarcinoma present at a younger age and with more risk factors in the late stage. These patients have significantly lower overall survival compared to their non-Hispanic counterparts. Hispanic patients in our study were 2.9 less likely to receive germline screening and more like to have somatic genetic actionable pathogenic variants. Overall, only a minority of all patients were enrolled in a pancreatic cancer clinical trial or offered genomic testing, highlighting a critical need and missed opportunity in advancing progress and improving outcomes for this disease, mainly in the underrepresented Hispanic population.
Collapse
Affiliation(s)
- Ritika Halder
- University of Arizona Internal Medicine Residency Program, Tucson, AZ, USA.
- Tecnológico de Monterrey Escuela de Medicina y Ciencias de la Salud, Monterrey, N.L., Mexico.
- University of Arizona Cancer Center, Tucson, AZ, USA.
| | - Sumana Veeravelli
- University of Arizona Internal Medicine Residency Program, Tucson, AZ, USA
- Tecnológico de Monterrey Escuela de Medicina y Ciencias de la Salud, Monterrey, N.L., Mexico
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Ce Cheng
- University of Arizona Internal Medicine Residency Program, Tucson, AZ, USA
- Tecnológico de Monterrey Escuela de Medicina y Ciencias de la Salud, Monterrey, N.L., Mexico
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Ricardo J Estrada-Mendizabal
- University of Arizona Internal Medicine Residency Program, Tucson, AZ, USA
- Tecnológico de Monterrey Escuela de Medicina y Ciencias de la Salud, Monterrey, N.L., Mexico
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Alejandro Recio-Boiles
- University of Arizona Internal Medicine Residency Program, Tucson, AZ, USA
- Tecnológico de Monterrey Escuela de Medicina y Ciencias de la Salud, Monterrey, N.L., Mexico
- University of Arizona Cancer Center, Tucson, AZ, USA
| |
Collapse
|
621
|
Hohmann N, Sprick MR, Pohl M, Ahmed A, Burhenne J, Kirchner M, Le Cornet L, Kratzmann M, Hajda J, Stenzinger A, Steindorf K, Delorme S, Schlemmer H, Riethdorf S, van Schaik R, Pantel K, Siveke J, Seufferlein T, Jäger D, Haefeli WE, Trumpp A, Springfeld C. Protocol of the IntenSify-Trial: An open-label phase I trial of the CYP3A inhibitor cobicistat and the cytostatics gemcitabine and nab-paclitaxel in patients with advanced stage or metastatic pancreatic ductal adenocarcinoma to evaluate the combination's pharmacokinetics, safety, and efficacy. Clin Transl Sci 2023; 16:2483-2493. [PMID: 37920921 PMCID: PMC10719473 DOI: 10.1111/cts.13661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 11/04/2023] Open
Abstract
Expression of CYP3A5 protein is a basal and acquired resistance mechanism of pancreatic ductal adenocarcinoma cells conferring protection against the CYP3A and CYP2C8 substrate paclitaxel through metabolic degradation. Inhibition of CYP3A isozymes restores the cells sensitivity to paclitaxel. The combination of gemcitabine and nab-paclitaxel is an established regimen for the treatment of metastasized or locally advanced inoperable pancreatic cancer. Cobicistat is a CYP3A inhibitor developed for the pharmacoenhancement of protease inhibitors. The addition of cobicistat to gemcitabine and nab-paclitaxel may increase the antitumor effect. We will conduct a phase I dose escalation trial with a classical 3 + 3 design to investigate the safety, tolerability, and pharmacokinetics (PKs) of gemcitabine, nab-paclitaxel, and cobicistat. Although the doses of gemcitabine (1000 mg/m2 ) and cobicistat (150 mg) are fixed, three dose levels of nab-paclitaxel (75, 100, and 125 mg/m2 ) will be explored to account for a potential PK drug interaction. After the dose escalation phase, we will set the recommended dose for expansion (RDE) and treat up to nine patients in an expansion part of the trial. The trial is registered under the following identifiers EudraCT-Nr. 2019-001439-29, drks.de: DRKS00029409, and ct.gov: NCT05494866. Overcoming resistance to paclitaxel by CYP3A5 inhibition may lead to an increased efficacy of the gemcitabine and nab-paclitaxel regimen. Safety, efficacy, PK, and RDE data need to be acquired before investigating this combination in a large-scale clinical study.
Collapse
Affiliation(s)
- Nicolas Hohmann
- Department of Medical OncologyHeidelberg University Hospital, National Center for Tumor DiseasesHeidelbergGermany
| | - Martin Ronald Sprick
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
| | - Moritz Pohl
- Institute of Medical BiometryUniversity Hospital HeidelbergHeidelbergGermany
| | - Azaz Ahmed
- Department of Medical OncologyHeidelberg University Hospital, National Center for Tumor DiseasesHeidelbergGermany
| | - Jürgen Burhenne
- Department of Clinical Pharmacology and PharmacoepidemiologyHeidelberg University HospitalHeidelbergGermany
| | - Marietta Kirchner
- Institute of Medical BiometryUniversity Hospital HeidelbergHeidelbergGermany
| | - Lucian Le Cornet
- NCT Trial Center, NCTGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Markus Kratzmann
- NCT Trial Center, NCTGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Jacek Hajda
- Pharmacovigilance Department, Coordination Centre for Clinical Trials (KKS)Heidelberg University HospitalHeidelbergGermany
| | | | - Karen Steindorf
- Division of Physical Activity, Prevention and CancerGerman Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) HeidelbergHeidelbergGermany
| | - Stefan Delorme
- Division of RadiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Sabine Riethdorf
- Institute of Tumor BiologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Ron van Schaik
- Department of Clinical ChemistryErasmus University Medical CenterRotterdamThe Netherlands
| | - Klaus Pantel
- Institute of Tumor BiologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Jens Siveke
- Bridge Institute of Experimental Tumor Therapy, West German Cancer CenterUniversity Hospital Essen, University of Duisburg‐EssenEssenGermany
- Division of Solid Tumor Translational OncologyGerman Cancer Consortium (DKTK Partner Site Essen) and German Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Dirk Jäger
- Department of Medical OncologyHeidelberg University Hospital, National Center for Tumor DiseasesHeidelbergGermany
- Clinical Cooperation Unit Applied Tumor ImmunityGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Walter E. Haefeli
- Department of Clinical Pharmacology and PharmacoepidemiologyHeidelberg University HospitalHeidelbergGermany
| | - Andreas Trumpp
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- German Cancer Consortium (DKTK)HeidelbergGermany
| | - Christoph Springfeld
- Department of Medical OncologyHeidelberg University Hospital, National Center for Tumor DiseasesHeidelbergGermany
| |
Collapse
|
622
|
van ‘t Land FR, Aziz MH, Michiels N, Mieog JSD, Bonsing BA, Luelmo SA, Homs MY, Groot Koerkamp B, Papageorgiou G, van Eijck CH. Increasing Systemic Immune-inflammation Index During Treatment in Patients With Advanced Pancreatic Cancer is Associated With Poor Survival: A Retrospective, Multicenter, Cohort Study. Ann Surg 2023; 278:1018-1023. [PMID: 37010512 PMCID: PMC10631500 DOI: 10.1097/sla.0000000000005865] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
BACKGROUND AND OBJECTIVES A high systemic immune-inflammation index (SIII) at diagnosis of various cancers, including pancreatic cancer, is associated with poor prognosis. The impact of FOLFIRINOX (5-fluorouracil, leucovorin, irinotecan, and oxaliplatin) chemotherapy or stereotactic body radiotherapy on this index is unknown. In addition, the prognostic value of changes in the SIII during treatment is unclear. In this retrospective analysis, we aimed to find answers regarding patients with advanced pancreatic cancer. METHODS Patients with advanced pancreatic cancer treated with FOLFIRINOX chemotherapy alone or with FOLFIRINOX chemotherapy followed by stereotactic body radiotherapy between 2015 and 2021 in 2 tertiary referral centers were included. Baseline characteristics, laboratory values at 3 time points during treatment, and survival outcomes were collected. The patient-specific evolutions of SIII and their association with mortality were assessed with joint models for longitudinal and time-to-event data. RESULTS Data of 141 patients were analyzed. At a median follow-up time of 23.0 months (95% CI: 14.6-31.3), 97 (69%) patients had died. Median overall survival was 13.2 months (95% CI: 11.0-15.5). During treatment with FOLFIRINOX, the log (SIII) was reduced by -0.588 (95% CI: -0.0978, -0.197; P = 0.003). One unit increase in log (SIII) increased the hazard ratio of dying by 1.604 (95% CI: 1.068-2.409; P = 0.023). CONCLUSIONS In addition to carbohydrate antigen 19-9, the SIII is a reliable biomarker in patients with advanced pancreatic cancer.
Collapse
Affiliation(s)
| | - Mohammad H. Aziz
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Nynke Michiels
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - J. Sven D. Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Bert A. Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Saskia A.C. Luelmo
- Department of Oncology, Leiden University Medical Center, the Netherlands
| | - Marjolein Y.V. Homs
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Grigorios Papageorgiou
- Department of Biostatistics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | | |
Collapse
|
623
|
Thalji SZ, Fernando D, Dua KS, Madhavan S, Chisholm P, Smith ZL, Aldakkak M, Christians KK, Clarke CN, George B, Kamgar M, Erickson BA, Hall WA, Evans DB, Tsai S. Biliary Adverse Events During Neoadjuvant Therapy for Pancreatic Cancer. Ann Surg 2023; 278:e1224-e1231. [PMID: 37078282 DOI: 10.1097/sla.0000000000005884] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
OBJECTIVE To describe a high-volume experience with biliary drainage before neoadjuvant therapy (NAT) for patients with operable pancreatic cancer (PC) and characterize the association between biliary adverse events (BAEs) and patient outcome. BACKGROUND Patients with PC presenting with biliary obstruction require durable decompression before NAT. METHODS Patients with operable PC and tumor-associated biliary obstruction were examined and grouped by the presence or absence of a BAE during NAT. The incidence, timing, and management of BAEs are described, and outcomes, including the completion of all treatment and overall survival (OS), were compared. RESULTS Of 426 patients who received pretreatment biliary decompression, 92 (22%) experienced at least 1 BAE during NAT, and 56 (13%) required repeat intervention on their biliary stent. The median duration of NAT was 161 days for all patients and was not different in the group that experienced BAEs. The median time from initial stent placement to BAE was 64 days. An interruption in the delivery of NAT (median 7 days) occurred in 25 (6%) of 426 patients. Among 426 patients, 290 (68%) completed all NAT, including surgery: 60 (65%) of 92 patients with BAE and 230 (69%) of 334 patients without BAE ( P =0.51). Among 290 patients who completed NAT and surgery, the median OS was 39 months, 26 months for the 60 patients with BAE, and 43 months for the 230 patients without BAE ( P =0.02). CONCLUSIONS During extended multimodal NAT for PC, 22% of patients experienced a BAE. Although BAEs were not associated with a significant interruption of treatment, patients who experienced a BAE had worse OS.
Collapse
Affiliation(s)
- Sam Z Thalji
- Division of Surgical Oncology, Department of Surgery, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Deemantha Fernando
- Division of Surgical Oncology, Department of Surgery, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Kulwinder S Dua
- Division of Gastroenterology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Srivats Madhavan
- Division of Gastroenterology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Phillip Chisholm
- Division of Gastroenterology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Zachary L Smith
- Division of Gastroenterology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Mohammed Aldakkak
- Division of Surgical Oncology, Department of Surgery, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Kathleen K Christians
- Division of Surgical Oncology, Department of Surgery, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Callisia N Clarke
- Division of Surgical Oncology, Department of Surgery, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Ben George
- Division of Medical Oncology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Mandana Kamgar
- Division of Medical Oncology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Beth A Erickson
- Department of Radiation Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - William A Hall
- Department of Radiation Oncology, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Douglas B Evans
- Division of Surgical Oncology, Department of Surgery, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| | - Susan Tsai
- Division of Surgical Oncology, Department of Surgery, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
624
|
Sugumar K, Hue JJ, Gupta S, Elshami M, Rothermel LD, Ocuin LM, Ammori JB, Hardacre JM, Winter JM. Trends in and Prognostic Significance of Time to Treatment in Pancreatic Cancer: A Population-Based Study. Ann Surg Oncol 2023; 30:8610-8620. [PMID: 37624518 DOI: 10.1245/s10434-023-14221-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 08/08/2023] [Indexed: 08/26/2023]
Abstract
INTRODUCTION The association of time to treatment (TTT) with survival remains unclear in pancreatic adenocarcinoma (PDAC). In this study, we evaluate the recent trends in TTT, causes for delay, and its effect on survival. METHODS We included patients with PDAC of all stages from the National Cancer Database (2004-2020) who underwent either surgery or chemotherapy/radiotherapy (CT/RT). TTT was defined as the duration between tissue diagnosis and first treatment. Linear regression (β) was used to study the temporal trends in time delay. RESULTS A total of 239,638 patients were included. The median TTT was 25 days. Using multivariable analysis, we found that increasing age (OR 1.48), female gender (OR 1.04), Black race (OR 1.3), lower educational status (OR 1.2), Medicaid, Medicare insurance, and uninsured (OR 1.2, 1.5, and 1.2, respectively), treatment at academic centers (OR 1.3), higher Charlson-Deyo comorbidity index (OR 1.2), and CT/RT (OR 1.5) were associated with increased TTT. There was a steady rise in median TTT from 21 to 28 days between 2004 and 2020 (β = 0.3), suggestive of a worsening trend. Concurrently, there was an increasing trend in utilization of neoadjuvant CT/RT between 2004 and 2020 in early-stage PDAC. On Cox regression, TTT delay was associated with poor overall survival in stage I-IV patients (HR 1.1, 1.1, 1.09, and 1.53, respectively). CONCLUSIONS Delayed treatment approaching 2 months was observed in 10% of the population. The rising temporal trend in TTT may be attributed to the increasing shift toward neoadjuvant CT/RT in early-stage PDAC and/or the increasing use of tissue biopsy prior to surgery.
Collapse
Affiliation(s)
- Kavin Sugumar
- Department of Surgery, University Hospitals Seidman Cancer Center and Case Comprehensive Cancer Center, University Hospitals Cleveland Medical, Cleveland, OH, USA.
| | - Jonathan J Hue
- Department of Surgery, University Hospitals Seidman Cancer Center and Case Comprehensive Cancer Center, University Hospitals Cleveland Medical, Cleveland, OH, USA
| | - Shreya Gupta
- Department of Surgery, University Hospitals Seidman Cancer Center and Case Comprehensive Cancer Center, University Hospitals Cleveland Medical, Cleveland, OH, USA
| | - Mohamedraed Elshami
- Department of Surgery, University Hospitals Seidman Cancer Center and Case Comprehensive Cancer Center, University Hospitals Cleveland Medical, Cleveland, OH, USA
| | - Luke D Rothermel
- Department of Surgery, University Hospitals Seidman Cancer Center and Case Comprehensive Cancer Center, University Hospitals Cleveland Medical, Cleveland, OH, USA
| | - Lee M Ocuin
- Department of Surgery, University Hospitals Seidman Cancer Center and Case Comprehensive Cancer Center, University Hospitals Cleveland Medical, Cleveland, OH, USA
| | - John B Ammori
- Department of Surgery, University Hospitals Seidman Cancer Center and Case Comprehensive Cancer Center, University Hospitals Cleveland Medical, Cleveland, OH, USA
| | - Jeffrey M Hardacre
- Department of Surgery, University Hospitals Seidman Cancer Center and Case Comprehensive Cancer Center, University Hospitals Cleveland Medical, Cleveland, OH, USA
| | - Jordan M Winter
- Department of Surgery, University Hospitals Seidman Cancer Center and Case Comprehensive Cancer Center, University Hospitals Cleveland Medical, Cleveland, OH, USA
| |
Collapse
|
625
|
Rasam S, Lin Q, Shen S, Straubinger RM, Qu J. Highly Reproducible Quantitative Proteomics Analysis of Pancreatic Cancer Cells Reveals Proteome-Level Effects of a Novel Combination Drug Therapy That Induces Cancer Cell Death via Metabolic Remodeling and Activation of the Extrinsic Apoptosis Pathway. J Proteome Res 2023; 22:3780-3792. [PMID: 37906173 DOI: 10.1021/acs.jproteome.3c00463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Pancreatic cancer patients have poor survival rates and are frequently treated using gemcitabine (Gem). However, initial tumor sensitivity often gives way to rapid development of resistance. Gem-based drug combinations are employed to increase efficacy and mitigate resistance, but our understanding of molecular-level drug interactions, which could assist in the development of more effective therapeutic regimens, is limited. Global quantitative proteomic analysis could provide novel mechanistic insights into drug combination interactions, but it is challenging to achieve high-quality quantitative proteomics analysis of the large sample sets that are typically required for drug combination studies. Here, we investigated molecular-level temporal interactions of Gem with BGJ398 (infigratinib), a recently approved pan-FGFR inhibitor, in multiple treatment groups (N = 42 samples) using IonStar, a robust large-scale proteomics method that employs well-controlled, ultrahigh-resolution MS1 quantification. A total of 5514 proteins in the sample set were quantified without missing data, requiring >2 unique peptides/protein, <1% protein false discovery rate (FDR), <0.1% peptide FDR, and CV < 10%. Functional analysis of the differentially altered proteins revealed drug-dysregulated processes such as metabolism, apoptosis, and antigen presentation pathways. These changes were validated experimentally using Seahorse metabolic assays and immunoassays. Overall, in-depth analysis of large-scale proteomics data provided novel insights into possible mechanisms by which FGFR inhibitors complement and enhance Gem activity in pancreatic cancers.
Collapse
Affiliation(s)
- Sailee Rasam
- Department of Biochemistry, University at Buffalo, Buffalo, New York 14260, United States
- New York State Center of Excellence in Bioinformatics and Life Sciences, 701 Ellicott Street, Buffalo, New York 14203, United States
| | - Qingxiang Lin
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York 14260, United States
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York 14203, United States
| | - Shichen Shen
- New York State Center of Excellence in Bioinformatics and Life Sciences, 701 Ellicott Street, Buffalo, New York 14203, United States
| | - Robert M Straubinger
- New York State Center of Excellence in Bioinformatics and Life Sciences, 701 Ellicott Street, Buffalo, New York 14203, United States
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York 14260, United States
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York 14203, United States
| | - Jun Qu
- Department of Biochemistry, University at Buffalo, Buffalo, New York 14260, United States
- New York State Center of Excellence in Bioinformatics and Life Sciences, 701 Ellicott Street, Buffalo, New York 14203, United States
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York 14260, United States
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York 14203, United States
| |
Collapse
|
626
|
Zhou K, Liu Y, Yuan S, Zhou Z, Ji P, Huang Q, Wen F, Li Q. Signalling in pancreatic cancer: from pathways to therapy. J Drug Target 2023; 31:1013-1026. [PMID: 37869884 DOI: 10.1080/1061186x.2023.2274806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
Pancreatic cancer (PC) is a common malignant tumour in the digestive system. Due to the lack of sensitive diagnostic markers, strong metastasis ability, and resistance to anti-cancer drugs, the prognosis of PC is inferior. In the past decades, increasing evidence has indicated that the development of PC is closely related to various signalling pathways. With the exploration of RAS-driven, epidermal growth factor receptor, Hedgehog, NF-κB, TGF-β, and NOTCH signalling pathways, breakthroughs have been made to explore the mechanism of pancreatic carcinogenesis, as well as the novel therapies. In this review, we discussed the signalling pathways involved in PC and summarised current targeted agents in the treatment of PC. Furthermore, opportunities and challenges in the exploration of potential therapies targeting signalling pathways were also highlighted.
Collapse
Affiliation(s)
- Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yingping Liu
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | | | - Ziyu Zhou
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Pengfei Ji
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Qianhan Huang
- School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Feng Wen
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
627
|
Kryh-Jensen CG, Fristrup CW, Ainsworth AP, Detlefsen S, Mortensen MB, Pfeiffer P, Tarpgaard LS, Graversen M. What is long-term survival in patients with peritoneal metastasis from gastric, pancreatic, or colorectal cancer? A study of patients treated with systemic chemotherapy and pressurized intraperitoneal aerosol chemotherapy (PIPAC). Pleura Peritoneum 2023; 8:147-155. [PMID: 38144215 PMCID: PMC10739291 DOI: 10.1515/pp-2023-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/27/2023] [Indexed: 12/26/2023] Open
Abstract
Objectives A definition of long-term survival (LTS) in patients with peritoneal metastasis (PM) from gastric cancer (GC), pancreatic cancer (PC) or colorectal cancer (CRC) treated with systemic chemotherapy and pressurized intraperitoneal aerosol chemotherapy (PIPAC) is lacking. We aimed to define LTS and investigate characteristics and treatment response in patients who reached LTS in data from two prospective trials. Methods Retrospective study of patients with GC-, PC-, or CRC-PM from the prospective PIPAC-OPC1 and PIPAC-OPC2 studies. The definition of LTS was based on published systematic reviews and randomized controlled trials. LTS was defined at the time point where 25 % of the patients were alive in these studies. Histology based response was evaluated by the mean Peritoneal Regression Grading Score (PRGS) using biopsies obtained prior to PIPAC 3, and defined by a mean PRGS of ≤2.0 or a decrease of mean PRGS of ≥1, compared to baseline. Results LTS was defined at 21 (GC), 15 (PC), and 24 (CRC) months. Fifty-one (47.2 %) patients (nine GC, 17 PC, 25 CRC) reached LTS calculated from the date of PM diagnosis. All but one received palliative chemotherapy before PIPAC, and 37 % received bidirectional treatment. More than 90 % of the LTS patients had response according to PRGS. The mOS from PIPAC 1 was 23.3, 12.4, and 28.5 months for GC, PC, and CRC LTS patients. Conclusions Patients with PM from GC, PC, and CRC treated with systemic chemotherapy and PIPAC can reach LTS and most show histological response. Causality must be further investigated.
Collapse
Affiliation(s)
- Charlotte G. Kryh-Jensen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Claus W. Fristrup
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Alan P. Ainsworth
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Sönke Detlefsen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Michael B. Mortensen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Per Pfeiffer
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Line S. Tarpgaard
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Martin Graversen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- OPEN – Odense Patient Data Explorative Network, Odense University Hospital, Region of Southern Denmark, Odense, Denmark
| |
Collapse
|
628
|
Xing J, Ge Y, Gong X, Liu Y, Cheng Y. Initial chemotherapy option for pancreatic ductal adenocarcinoma in patients with adequate performance status. JOURNAL OF PANCREATOLOGY 2023; 6:196-201. [DOI: 10.1097/jp9.0000000000000144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly progressive lethal malignancy, with chemotherapy being the primary treatment modality. This article provides a review of the initial chemotherapy options for PDAC patients with adequate performance status, comparing FOLFIRINOX (oxaliplatin, irinotecan, 5-fluorouracil, and leucovorin) or modified FOLFIRINOX and gemcitabine plus nab-paclitaxel (GEM-NabP) regimens. The availability of limited evidence from randomized trials restricts a direct comparison between the 2 regimens. Based on our review, (m)FOLFIRINOX yields superior survival outcomes compared to GEM-NabP in metastatic PDAC. For locally advanced PDAC, either (m)FOLFIRINOX or GEM-NabP can be considered initial chemotherapy. In the neoadjuvant setting for borderline resectable PDAC, both regimens have demonstrated promising results in achieving feasible resection rates. However, mFOLFIRINOX remains the preferred choice for adjuvant chemotherapy. The selection of initial chemotherapy for PDAC depends on the disease stage, patients’ performance status, and tumor molecular alterations. Further research and clinical trials are necessary to optimize treatment approaches for PDAC patients.
Collapse
Affiliation(s)
- Jiazhang Xing
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuping Ge
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaolei Gong
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Liu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
629
|
Berger JM, Alany A, Berchtold L, Puhr R, Friedrich A, Scheiner B, Prager GW, Preusser M, Berghoff AS, Bergen ES. Prognosticators of survival in patients with metastatic pancreatic cancer and ascites. ESMO Open 2023; 8:102048. [PMID: 37977000 PMCID: PMC10774951 DOI: 10.1016/j.esmoop.2023.102048] [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: 08/02/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Identification of factors associated with survival after ascites diagnosis in metastatic pancreatic cancer (mPC) patients may guide treatment decisions and help to maintain quality of life in this highly symptomatic patient collective. PATIENTS AND METHODS All patients treated for mPC at the Medical University of Vienna between 2010 and 2019 developing ascites throughout their course of disease were identified by retrospective chart review. General risk factors, metastatic sites, systemic inflammation and liver function parameters, as well as type of treatment after ascites diagnosis were investigated for associations with survival. RESULTS One hundred and seventeen mPC patients with ascites were included in this study. Median time from mPC to ascites diagnosis was 8.9 months (range 0-99 months) and median overall survival (OS) after ascites diagnosis was 27.4 days (range 21.3-42.6 days). Identified prognostic factors at ascites diagnosis independently associated with an impaired OS were presence of liver metastases [hazard ratio (HR): 2.07, 95% confidence interval (CI) 1.13-3.79, P = 0.018), peritoneal carcinomatosis (HR: 1.74, 95% CI 1.11-2.71, P = 0.015), and portal vein obstruction (HR: 2.52, 95% CI 1.29-4.90, P = 0.007). Compared with best supportive care, continuation of systemic therapy after ascites diagnosis was independently associated with survival (HR: 0.35, 95% CI 0.20-0.61, P < 0.001) with a median OS of 62 days (95% CI 51-129 days, P < 0.001) versus 16 days (95% CI 11-24 days), respectively. CONCLUSIONS Liver and peritoneal metastases as well as portal vein obstruction were found to be prognostic factors after ascites diagnosis in mPC patients. Continuation of systemic therapy after ascites diagnosis was associated with a longer OS, which needs to be evaluated in larger clinical trials including quality-of-life assessment.
Collapse
Affiliation(s)
- J M Berger
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna
| | - A Alany
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - L Berchtold
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - R Puhr
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - A Friedrich
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - B Scheiner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - G W Prager
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - M Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna
| | - A S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna
| | - E S Bergen
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna.
| |
Collapse
|
630
|
Xiang F, Luo F. Stem cell factor modulates HIF-1α levels and diminishes 5-FU sensitivity in 5-FU resistant pancreatic cells by altering the anabolic glucose metabolism. J Biochem Mol Toxicol 2023; 37:e23487. [PMID: 37718545 DOI: 10.1002/jbt.23487] [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: 03/30/2023] [Revised: 06/19/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023]
Abstract
Resistance to chemotherapy in cancer leads to poor therapeutic outcomes and also leads to challenges in treatment. The present work evaluated the mechanism involved in the resistance of 5-flurouracil (5-FU) in pancreatic cancer. At least 14 different pancreatic cancer (PC) cell lines were used for the study. For in vivo study female nude mice were selected. Patient-derived tumor xenograft samples were obtained from patients. The study involved, study for glucose uptake, fluorescence-activated cell sorting for glucose transporter, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide for cell survival, Picto-micrography for clonogenic assay, glutamine uptake assay, extracellular acidification and oxygen consumption rate, carbon dioxide release assay and lactate assay were also done. In addition to this, quantitative real-time polymerase chain reaction analysis for expression of genes, chromatin immunoprecipitation assay, western blot for protein expression, and immunohistochemical analysis in tumor sections, the tumors were studied by imaging for hypoxia and localization of TKT and CTPS-2. Also, patient-derived xenograft tumors were engrafted in nude mice, followed by a glucose uptake assay. We reported that elevated glycolytic flux causes dependence on glucose in cancer cells and, at the same time, increases pyrimidine biosynthesis. It was also found that stem cell factor-mediated stabilization of hypoxia-inducible factor-1a (HIF-1α) modulates the resistance in PC. Targeting HIF-1α in combination with 5-FU, strongly reduced the tumor burden. The study concludes that stem cell factor modulates HIF-1α and decreases the sensitivity in 5-FU resistant pancreatic cancer cells by targeting glucose metabolism. Deceased expression levels of CTPS-2 and TKT, which are regulators of pyrimidine biosynthesis could better the chance of survival in patients of pancreatic cancer receiving treatment of 5-FU.
Collapse
Affiliation(s)
- Fu Xiang
- Department of General Surgery, Dalian Medical University, Dalian, Liaoning, China
| | - Fuwen Luo
- Department of Acute Abdominal Surgery, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
631
|
Mohamed A, Nicolais L, Fitzgerald TL. Pancreatic Cancer with Vascular Involvement: Adherence to Current Standard-of-Care Associated with Improved Survival. Am Surg 2023; 89:5535-5544. [PMID: 36854081 DOI: 10.1177/00031348231156756] [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] [Indexed: 03/02/2023]
Abstract
METHODS This study is a retrospective cohort study of National Cancer Data Base (NCDB) data for pancreatic cancer with vascular involvement. RESULTS A total of 23 903 patients with vascular involvement were included and divided into 3 groups; no treatment (40.6%), medical treatment (36.6%), and resection (22.8%). Of the patients undergoing resection, 31.3% received neoadjuvant multiagent chemotherapy (N-MAC). The remainder were treated with postoperative adjuvant treatment (33.8%), surgery alone (24.9%), preoperative radiotherapy (8.3%), or single-agent preoperative chemotherapy (1.7%). Median survival for N-MAC was superior (28.42 months) when compared to neoadjuvant radiotherapy (20.73 months), neoadjuvant single-agent chemotherapy (20.8 months), postoperative adjuvant therapy (17.87 months), and surgery alone (10.12 months). N-MAC was associated with improved survival compared to postoperative multiagent chemotherapy (P-MAC) (28.4 vs 16.95, HR 1.82; CI 1.64-2.02, P < .0010) (Figure 1). The addition of radiation therapy to N-MAC did not improve survival (27.4 vs 29.8, HR .93; CI .83-1.05, P = .3). Clinical downstaging occurred in 40% of patients treated with N-MAC, and downstaging was associated with improved survival (HR .74; CI .64-.85, P < .001). N-MAC patients were more likely to undergo an R0 resection than P-MAC (74% v. 48, P < .001). CONCLUSIONS Most resected pancreatic cancer patients in this study with vascular involvement receive either postoperative or no adjuvant therapy. N-MAC increases downstaging, R0 resection rates, and survival.
Collapse
Affiliation(s)
- Abdimajid Mohamed
- Division of Surgical Oncology, Tufts University School of Medicine, Boston, MA, USA
| | | | - Timothy L Fitzgerald
- Division of Surgical Oncology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
632
|
Shimoyama R, Imamura Y, Uryu K, Mase T, Fujimura Y, Hayashi M, Ohtaki M, Ohtani K, Shinozaki N, Minami H. Real‑world treatment outcomes among patients with metastatic pancreatic cancer in Japan: The Tokushukai real‑world data project. Mol Clin Oncol 2023; 19:98. [PMID: 37953858 PMCID: PMC10636700 DOI: 10.3892/mco.2023.2694] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/07/2023] [Indexed: 11/14/2023] Open
Abstract
The present study aimed to investigate temporal trends in treatment patterns and prognostic factors for overall survival (OS) among patients with metastatic pancreatic cancer. From the Tokushukai REAl-world Data project, 1,093 patients with metastatic pancreatic cancer treated with gemcitabine, tegafur/gimeracil/oteracil (S-1), gemcitabine plus S-1, gemcitabine plus nab-paclitaxel, or fluorouracil, folic acid, oxaliplatin and irinotecan (FOLFIRINOX) between April 2010 and March 2020 were identified. Stratified/conventional Cox regression analyses were conducted to examine associations between patient- and tumor-related factors, study period, hospital volume, hospital type and first-line chemotherapy regimens. Overall, 846 patients were selected (503 male patients; median age, 70 years) after excluding ineligible patients. Over a median follow-up of 5.4 months, the median OS was 6.8 months (95% confidence interval, 6.3-7.4). The median OS for gemcitabine, S-1, gemcitabine plus S-1, gemcitabine plus nab-paclitaxel and FOLFIRINOX regimens was 5.9, 5.3, 7.7, 9.0 and 9.5 months, respectively. The median OS for 2010-2013, 2014-2017 and 2017-2020 was 6.2, 7.1 and 7.8 months, respectively. Performance status, body mass index and first-line chemotherapy regimens were identified to be significant prognostic factors. In summary, the real-world data indicated that standard care, including chemotherapy, for metastatic pancreatic cancer was widely used in hospitals throughout Japan and verified the survival benefits of gemcitabine plus nab-paclitaxel and FOLFIRINOX observed in prior clinical trials. This trial has been registered in the University Hospital Medical Information Network Clinical Trials Registry as UMIN000050590 on April 1, 2023.
Collapse
Affiliation(s)
- Rai Shimoyama
- Department of General Surgery, Shonan Kamakura General Hospital, Kamakura, Kanagawa 247-8533, Japan
| | - Yoshinori Imamura
- Department of Medical Oncology and Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Kiyoaki Uryu
- Department of Medical Oncology, Yao Tokushukai General Hospital, Yao, Osaka 581-0011, Japan
| | - Takahiro Mase
- Department of Breast Surgery, Ogaki Tokushukai Hospital, Ogaki, Gifu 503-0015, Japan
| | | | - Maki Hayashi
- Mirai Iryo Research Center Inc., Tokyo 102-0074, Japan
| | - Megu Ohtaki
- deCult Co., Ltd., Hatsukaichi, Hiroshima 739-0413, Japan
| | - Keiko Ohtani
- deCult Co., Ltd., Hatsukaichi, Hiroshima 739-0413, Japan
| | - Nobuaki Shinozaki
- Department of General Surgery, Shonan Kamakura General Hospital, Kamakura, Kanagawa 247-8533, Japan
| | - Hironobu Minami
- Department of Medical Oncology and Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
- Cancer Center, Kobe University Hospital, Kobe, Hyogo 650-0017, Japan
| |
Collapse
|
633
|
Militello AM, Orsi G, Cavaliere A, Niger M, Avallone A, Salvatore L, Tortora G, Rapposelli IG, Giordano G, Noventa S, Giommoni E, Bozzarelli S, Macchini M, Peretti U, Procaccio L, Puccini A, Cascinu S, Montagna C, Milella M, Reni M. Clinical outcomes and response to chemotherapy in a cohort of pancreatic cancer patients with germline variants of unknown significance (VUS) in BRCA1 and BRCA2 genes. Cancer Chemother Pharmacol 2023; 92:501-510. [PMID: 37725113 DOI: 10.1007/s00280-023-04585-w] [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: 02/05/2023] [Accepted: 08/22/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE The clinical outcome and the efficacy of chemotherapy in pancreatic cancer patients with BRCA1/2 Variants of Unknown Significance (VUS) is unknown. We explored the effects of chemotherapy with or without Platinum in non metastatic and metastatic pancreatic cancer patients with BRCA1/2 VUS. METHODS A retrospective analysis of non-metastatic or metastatic pancreatic cancer patients with gBRCA1/2 VUS treated in 13 Italian centers between November 2015 and December 2020 was performed. All patients were assessed for toxicity and RECIST 1.1 response. Metastatic patients were evaluated for survival outcome. RESULTS 30 pancreatic cancer patients with gBRCA1/2 VUS were considered: 20 were M+ and 10 were non-M+. Pl-CT was recommended to 16 patients: 10 M+ (6 FOLFIRINOX and 4 PAXG) and 6 non-M+ (3 FOLFIRINOX and 3 PAXG); 11 patients received Nabpaclitaxel-Gemcitabine (AG; 8 M+) and 3 patients (2 M+) were treated with Gemcitabine (G). The RECIST 1.1 response rate was 27% for AG and 44% for Pl-CT (22% for (m) FOLFIRINOX and 71% PAXG). 1 year Progression-Free Survival was 37.5% for patients treated with AG and 33% in the Pl-CT subgroup. Median Overall Survival (OS) was 23.5 months for patients treated with AG and 14 months for the Pl-CT subgroup. 1 Year and 2 Year OS were numerically better for AG (1 Year OS: 75% vs 60% and 2 Year OS: 50% and 20% in AG and Pl-CT subgroups, respectively) as well. CONCLUSIONS Pl-CT does not seem to be associated with a better outcome compared to AG chemotherapy in PDAC patients with BRCA 1/2 VUS.
Collapse
Affiliation(s)
- Anna Maria Militello
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Orsi
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Cavaliere
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO - IRCCS Candiolo, Candiolo, Italy
| | - Monica Niger
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Antonio Avallone
- Biologia Cellulare e Bioterapie, Istituto Nazionale per lo Studio e la Cura dei Tumori ''Fondazione Giovanni Pascale'' - IRCCS, Naples, Italy
| | - Lisa Salvatore
- Unit of Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario, Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giampaolo Tortora
- Unit of Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario, Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ilario Giovanni Rapposelli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ''Dino Amadori'', Meldola, Italy
| | - Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy, Policlinico Riuniti, Foggia, Italy
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Silvia Noventa
- Department of Medical Oncology, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Elisa Giommoni
- Medical Oncology Division, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Silvia Bozzarelli
- Department of Medical Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Marina Macchini
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Umberto Peretti
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Letizia Procaccio
- Medical Oncology 1 Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Alberto Puccini
- University of Genoa, Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Stefano Cascinu
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Montagna
- Department of Radiation Oncology and Genomic Instability and Cancer Genetics, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Michele Reni
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy.
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
634
|
Gasparini G, Aleotti F, Palucci M, Belfiori G, Tamburrino D, Partelli S, Orsi G, Macchini M, Archibugi L, Capurso G, Arcidiacono PG, Crippa S, Reni M, Falconi M. The role of biliary events in treatment and survival of patients with advanced pancreatic ductal adenocarcinoma. Dig Liver Dis 2023; 55:1750-1756. [PMID: 37121820 DOI: 10.1016/j.dld.2023.04.015] [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: 11/25/2022] [Revised: 04/09/2023] [Accepted: 04/13/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND Systemic chemotherapy (CT) is the treatment of choice for advanced pancreatic ductal adenocarcinoma (PDAC). Biliary obstruction is common in this setting and may interfere with CT administration due to jaundice or cholangitis related to biliary stent malfunction. AIMS To evaluate the impact of biliary events on CT administration and survival in patients with stage III-IV PDAC. METHODS Patients enrolled in a randomized trial of nab-paclitaxel plus gemcitabine with/without capecitabine and cisplatin in advanced PDAC were included. Data on management of jaundice, biliary stents/complications and CT were prospectively collected and retrospectively analyzed. Modified overall (mOS) and progression-free (mPFS) survival were evaluated. RESULTS Eighty-eight patients met the inclusion criteria (50% females; median age 65years). Seven of eight (87.5%) patients who placed plastic stents developed biliary complications versus 14/30 (46.7%) with metallic stents (p = 0.071). Patients without biliary complications completed planned CT in 64.2% versus 47.6% of cases (p = 0.207). CT completion was related to longer mOS (17 vs 12 months, p = 0.005) and mPFS (9 vs 6 months, p = 0.011). mOS was shorter when biliary complications occurred (12 vs 17 months, p = 0.937), as was mPFS (6 vs 8 months, p = 0.438). CONCLUSION Complications related to biliary obstruction influence chemotherapy completion and survival in patients with advanced PDAC.
Collapse
Affiliation(s)
- Giulia Gasparini
- Pancreatic and Transplant Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Aleotti
- Pancreatic and Transplant Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Palucci
- Pancreatic and Transplant Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Belfiori
- Pancreatic and Transplant Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Domenico Tamburrino
- Pancreatic and Transplant Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Partelli
- Pancreatic and Transplant Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy, Vita-Salute San Raffaele University, Milan, Italy
| | - Giulia Orsi
- Division of Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marina Macchini
- Division of Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Livia Archibugi
- Division of Pancreato-biliary Endoscopy and Endosonography, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gabriele Capurso
- Division of Pancreato-biliary Endoscopy and Endosonography, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy, Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Giorgio Arcidiacono
- Division of Pancreato-biliary Endoscopy and Endosonography, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy, Vita-Salute San Raffaele University, Milan, Italy
| | - Stefano Crippa
- Pancreatic and Transplant Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy, Vita-Salute San Raffaele University, Milan, Italy.
| | - Michele Reni
- Division of Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy, Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Falconi
- Pancreatic and Transplant Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
635
|
Yu H, Sercel ZP, Rezgui SP, Farhi J, Virgil SC, Stoltz BM. Total Synthesis of Aleutianamine. J Am Chem Soc 2023; 145:25533-25537. [PMID: 37967164 PMCID: PMC10690800 DOI: 10.1021/jacs.3c10212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/17/2023]
Abstract
Aleutianamine is a recently isolated pyrroloiminoquinone natural product that displays potent and selective biological activity toward human pancreatic cancer cells with an IC50 of 25 nM against PANC-1, making it a potential candidate for therapeutic development. We report a synthetic approach to aleutianamine wherein the unique [3.3.1] ring system and tertiary sulfide of this alkaloid were constructed via a novel palladium-catalyzed dearomative thiophene functionalization. Other highlights of the synthesis include a palladium-catalyzed decarboxylative pinacol-type rearrangement of an allylic carbonate to install a ketone and a late-stage oxidative amination. This concise and convergent strategy will enable access to analogues of aleutianamine and further investigation of the biological activity of this unique natural product.
Collapse
Affiliation(s)
| | | | - Samir P. Rezgui
- The Warren and Katharine
Schlinger Laboratory for Chemistry and Chemical Engineering, Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Jonathan Farhi
- The Warren and Katharine
Schlinger Laboratory for Chemistry and Chemical Engineering, Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Scott C. Virgil
- The Warren and Katharine
Schlinger Laboratory for Chemistry and Chemical Engineering, Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Brian M. Stoltz
- The Warren and Katharine
Schlinger Laboratory for Chemistry and Chemical Engineering, Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
636
|
Zheng Q, Tang J, Aicher A, Bou Kheir T, Sabanovic B, Ananthanarayanan P, Reina C, Chen M, Gu JM, He B, Alcala S, Behrens D, Lawlo RT, Scarpa A, Hidalgo M, Sainz B, Sancho P, Heeschen C. Inhibiting NR5A2 targets stemness in pancreatic cancer by disrupting SOX2/MYC signaling and restoring chemosensitivity. J Exp Clin Cancer Res 2023; 42:323. [PMID: 38012687 PMCID: PMC10683265 DOI: 10.1186/s13046-023-02883-y] [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: 08/09/2023] [Accepted: 11/02/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a profoundly aggressive and fatal cancer. One of the key factors defining its aggressiveness and resilience against chemotherapy is the existence of cancer stem cells (CSCs). The important task of discovering upstream regulators of stemness that are amenable for targeting in PDAC is essential for the advancement of more potent therapeutic approaches. In this study, we sought to elucidate the function of the nuclear receptor subfamily 5, group A, member 2 (NR5A2) in the context of pancreatic CSCs. METHODS We modeled human PDAC using primary PDAC cells and CSC-enriched sphere cultures. NR5A2 was genetically silenced or inhibited with Cpd3. Assays included RNA-seq, sphere/colony formation, cell viability/toxicity, real-time PCR, western blot, immunofluorescence, ChIP, CUT&Tag, XF Analysis, lactate production, and in vivo tumorigenicity assays. PDAC models from 18 patients were treated with Cpd3-loaded nanocarriers. RESULTS Our findings demonstrate that NR5A2 plays a dual role in PDAC. In differentiated cancer cells, NR5A2 promotes cell proliferation by inhibiting CDKN1A. On the other hand, in the CSC population, NR5A2 enhances stemness by upregulating SOX2 through direct binding to its promotor/enhancer region. Additionally, NR5A2 suppresses MYC, leading to the activation of the mitochondrial biogenesis factor PPARGC1A and a shift in metabolism towards oxidative phosphorylation, which is a crucial feature of stemness in PDAC. Importantly, our study shows that the specific NR5A2 inhibitor, Cpd3, sensitizes a significant fraction of PDAC models derived from 18 patients to standard chemotherapy. This treatment approach results in durable remissions and long-term survival. Furthermore, we demonstrate that the expression levels of NR5A2/SOX2 can predict the response to treatment. CONCLUSIONS The findings of our study highlight the cell context-dependent effects of NR5A2 in PDAC. We have identified a novel pharmacological strategy to modulate SOX2 and MYC levels, which disrupts stemness and prevents relapse in this deadly disease. These insights provide valuable information for the development of targeted therapies for PDAC, offering new hope for improved patient outcomes. A Schematic illustration of the role of NR5A2 in cancer stem cells versus differentiated cancer cells, along with the action of the NR5A2 inhibitor Cpd3. B Overall survival of tumor-bearing mice following allocated treatment. A total of 18 PDX models were treated using a 2 x 1 x 1 approach (two animals per model per treatment); n=36 per group (illustration created with biorender.com ).
Collapse
Affiliation(s)
- Quan Zheng
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajia Tang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Alexandra Aicher
- Precision Immunotherapy, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Immunology Research and Development Center, China Medical University, Taichung, Taiwan
| | - Tony Bou Kheir
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Berina Sabanovic
- Pancreatic Cancer Heterogeneity Lab, Candiolo Cancer Institute - FPO - IRCCS, Candiolo, Turin, Italy
| | - Preeta Ananthanarayanan
- Pancreatic Cancer Heterogeneity Lab, Candiolo Cancer Institute - FPO - IRCCS, Candiolo, Turin, Italy
| | - Chiara Reina
- Pancreatic Cancer Heterogeneity Lab, Candiolo Cancer Institute - FPO - IRCCS, Candiolo, Turin, Italy
| | - Minchun Chen
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Min Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Sonia Alcala
- Molecular Pathology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Chronic Diseases and Cancer Area 3 Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red, Área Cáncer, CIBERONC, ISCIII, Madrid, Spain
| | - Diana Behrens
- Experimental Pharmacology and Oncology Berlin-Buch GmbH, Berlin, Germany
| | - Rita T Lawlo
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
- ARC-Net, Applied Research On Cancer Centre, University of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
- ARC-Net, Applied Research On Cancer Centre, University of Verona, Verona, Italy
| | - Manuel Hidalgo
- Clinical Research Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Bruno Sainz
- Molecular Pathology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Chronic Diseases and Cancer Area 3 Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red, Área Cáncer, CIBERONC, ISCIII, Madrid, Spain
| | - Patricia Sancho
- IIS Aragon, Hospital Universitario Miguel Servet, 50009, Saragossa, Spain.
| | - Christopher Heeschen
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Pancreatic Cancer Heterogeneity Lab, Candiolo Cancer Institute - FPO - IRCCS, Candiolo, Turin, Italy.
- Molecular Pathology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| |
Collapse
|
637
|
Augustinus S, van Laarhoven HWM, Cirkel GA, de Groot JWB, Groot Koerkamp B, Macarulla T, Melisi D, O’Reilly EM, van Santvoort HC, Mackay TM, Besselink MG, Wilmink JW. Timing of Initiation of Palliative Chemotherapy in Asymptomatic Patients with Metastatic Pancreatic Cancer: An International Expert Survey and Case-Vignette Study. Cancers (Basel) 2023; 15:5603. [PMID: 38067306 PMCID: PMC10705283 DOI: 10.3390/cancers15235603] [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: 09/19/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 02/15/2024] Open
Abstract
Background: The use of imaging, in general, and during follow-up after resection of pancreatic cancer, is increasing. Consequently, the number of asymptomatic patients diagnosed with metastatic pancreatic cancer (mPDAC) is increasing. In these patients, palliative systemic therapy is the only tumor-directed treatment option; hence, it is often immediately initiated. However, delaying therapy in asymptomatic palliative patients may preserve quality of life and avoid therapy-related toxicity, but the impact on survival is unknown. This study aimed to gain insight into the current perspectives and clinical decision=making of experts regarding the timing of treatment initiation of patients with asymptomatic mPDAC. Methods: An online survey (13 questions, 9 case-vignettes) was sent to all first and last authors of published clinical trials on mPDAC over the past 10 years and medical oncologists of the Dutch Pancreatic Cancer Group. Inter-rater variability was determined using the Kappa Light test. Differences in the preferred timing of treatment initiation among countries, continents, and years of experience were analyzed using Fisher's exact test. Results: Overall, 78 of 291 (27%) medical oncologists from 15 countries responded (62% from Europe, 23% from North America, and 15% from Asia-Pacific). The majority of respondents (63%) preferred the immediate initiation of chemotherapy following diagnosis. In 3/9 case-vignettes, delayed treatment was favored in specific clinical contexts (i.e., patient with only one small lung metastasis, significant comorbidities, and higher age). A significant degree of inter-rater variability was present within 7/9 case-vignettes. The recommended timing of treatment initiation differed between continents for 2/9 case-vignettes (22%), in 7/9 (77.9%) comparing the Netherlands with other countries, and based on years of experience for 5/9 (56%). Conclusions: Although the response rate was limited, in asymptomatic patients with mPDAC, immediate treatment is most often preferred. Delaying treatment until symptoms occur is considered in patients with limited metastatic disease, more comorbidities, and higher age.
Collapse
Affiliation(s)
- Simone Augustinus
- Department of Surgery, Location University of Amsterdam, Amsterdam University Medical Center, 1105 Amsterdam, The Netherlands; (S.A.); (M.G.B.)
- Cancer Center Amsterdam, 1105 Amsterdam, The Netherlands
| | - Hanneke W. M. van Laarhoven
- Cancer Center Amsterdam, 1105 Amsterdam, The Netherlands
- Department of Medical Oncology, Location University of Amsterdam, Amsterdam University Medical Center, 1105 Amsterdam, The Netherlands
| | - Geert A. Cirkel
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, 3584 Utrecht, The Netherlands
| | | | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, 3015 Rotterdam, The Netherlands
| | - Teresa Macarulla
- Department of Medical Oncology, Vall d’Hebron Unveristy Hospital, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Davide Melisi
- Digestive Molecular Clinical Oncology Unit, Univeristy of Verona, 37134 Verona, Italy
| | - Eileen M. O’Reilly
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hjalmar C. van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, St. Antonius Hospital Nieuwegein, Utrecht University, 3584 Utrech, The Netherlands
| | - Tara M. Mackay
- Department of Surgery, Location University of Amsterdam, Amsterdam University Medical Center, 1105 Amsterdam, The Netherlands; (S.A.); (M.G.B.)
- Cancer Center Amsterdam, 1105 Amsterdam, The Netherlands
| | - Marc G. Besselink
- Department of Surgery, Location University of Amsterdam, Amsterdam University Medical Center, 1105 Amsterdam, The Netherlands; (S.A.); (M.G.B.)
- Cancer Center Amsterdam, 1105 Amsterdam, The Netherlands
| | - Johanna W. Wilmink
- Cancer Center Amsterdam, 1105 Amsterdam, The Netherlands
- Department of Medical Oncology, Location University of Amsterdam, Amsterdam University Medical Center, 1105 Amsterdam, The Netherlands
| |
Collapse
|
638
|
Schroeder BA, Mandelson MT, Picozzi VJ. Alternating Gemcitabine/Nab-Paclitaxel (GA) and 5-FU/Leucovorin/Irinotecan (FOLFIRI) as First-Line Treatment for De Novo Metastatic Pancreatic Cancer (MPC): Safety and Effect. Cancers (Basel) 2023; 15:5588. [PMID: 38067292 PMCID: PMC10705182 DOI: 10.3390/cancers15235588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/01/2023] [Accepted: 11/23/2023] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Both gemcitabine- and 5-fluorouracil (5-FU)-based chemotherapy regimens have demonstrated efficacy in metastatic pancreatic cancer (MPC). Alternating these regimens may reduce toxicity, slow resistant cancer biology emergence, and provide a platform for the addition of other therapeutic agents. Alternating gemcitabine/nab-paclitaxel (GA) and 5-FU/leucovorin/irinotecan (FOLFIRI) in MPC has previously been reported at our own institution and elsewhere. An extension of our institutional observations is reported here. METHODS Patient eligibility required the following: biopsy-proven de novo MPC, no prior evidence of disease on CT, ECOG performance status (PS) ≤ 2, and bi-dimensionally measurable disease. Treatment (Tx) entailed gemcitabine 1000 mg/m2 and nab-paclitaxel 125 mg/m2 1, (8), 15 alternating every 8 weeks (2 cycles) with FOLFIRI using standard dosing. Patients were radiographically re-staged every 8 weeks. Tx spanned up to 12 cycles. Tx thereafter was decided following patient/physician discussion. RESULTS Median overall survival (mOS) was 13.2 months (95% CI 10.9-16.5 months). Median progression-free survival (mPFS) was 8.5 months (95% CI, 7.1-9.9). The 6-, 12-, 18-, and 24-month OS rates were 88%, 54%, 36%, and 20%, respectively. The disease control rate at 16 weeks was 83% (37% PR, 46% SD). Hematologic toxicity grade ≥ 3 included 9.3% anemia, 10.2% neutropenia, and 4.6% thrombocytopenia. Neutrophil growth factors were not used in this cohort. Non-hematologic toxicities grade ≥ 3 included neuropathy 0.9%, nausea/vomiting 0.9%, and diarrhea 0.9%. No patients experienced mucositis on this regimen. CONCLUSIONS Alternating GA/FOLFIRI in MPC has a favorable toxicity profile in comparison to current standard regimens. Median OS was at least competitive with standard regimens, and longer-term (18 and 24 months) OS seemed particularly encouraging. Treatment for ≥48 weeks and ECOG PS of zero at the time of treatment initiation were prognostically significant. Further investigation using this regimen including randomized comparisons, the incorporation of molecular data, and use of additional agents is merited.
Collapse
Affiliation(s)
- Brett A. Schroeder
- Virginia Mason Medical Center, Seattle, WA 98101, USA; (M.T.M.); (V.J.P.)
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Margaret T. Mandelson
- Virginia Mason Medical Center, Seattle, WA 98101, USA; (M.T.M.); (V.J.P.)
- Benaroya Research Institute, Seattle, WA 98101, USA
| | - Vincent J. Picozzi
- Virginia Mason Medical Center, Seattle, WA 98101, USA; (M.T.M.); (V.J.P.)
| |
Collapse
|
639
|
Qin S, Li J, Bai Y, Wang Z, Chen Z, Xu R, Xu J, Zhang H, Chen J, Yuan Y, Liu T, Yang L, Zhong H, Chen D, Shen L, Hao C, Fu D, Cheng Y, Yang J, Wang Q, Qin B, Pan H, Zhang J, Bai X, Zheng Q. Nimotuzumab Plus Gemcitabine for K-Ras Wild-Type Locally Advanced or Metastatic Pancreatic Cancer. J Clin Oncol 2023; 41:5163-5173. [PMID: 37647576 PMCID: PMC10666986 DOI: 10.1200/jco.22.02630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/19/2023] [Accepted: 07/20/2023] [Indexed: 09/01/2023] Open
Abstract
PURPOSE In a phase IIb trial of nimotuzumab plus gemcitabine, substantial clinical benefits were observed in patients with locally advanced or metastatic pancreatic cancer (PC). Therefore, we conducted a phase III clinical study to verify the efficacy and safety of this combination regimen in patients with K-Ras wild-type tumors (ClinicalTrials.gov identifier: NCT02395016). PATIENTS AND METHODS Eligible patients were randomly assigned to receive nimotuzumab (400 mg once per week) or placebo followed by gemcitabine (1,000 mg/m2 on days 1, 8, and 15, once every 4 weeks) until disease progression or unacceptable toxicity. The primary end point was overall survival (OS) and the secondary end points were progression-free survival (PFS), response rates, and safety. RESULTS A total of 480 patients were screened; 92 patients were enrolled and 82 patients with K-Ras wild-type tumors were eligible. In the full analysis set, the median OS was 10.9 versus 8.5 months, while the restricted mean survival time (RMST) was 18.05 versus 11.14 months for the investigational versus control arm (ratio of control v investigation = 0.62 [0.40-0.97]; P = .036). Median PFS was 4.2 versus 3.6 months in the investigational versus control arm (log-rank P = .04; hazard ratio, 0.60 [0.37-0.99]) and the restricted mean PFS time was 8.08 versus 4.76 months (RMST ratio, 0.58 [0.38-0.90]; P = .036). Both OS and PFS were longer in the nimotuzumab group than in the placebo group. The objective response rates and disease control rates were 7% versus 10% and 68% versus 63% for the investigational and control groups, respectively. The incidence of adverse events were comparable between the two groups. CONCLUSION In patients with locally advanced or metastatic K-Ras wild-type PC, nimotuzumab plus gemcitabine significantly improved OS and PFS with a good safety profile.
Collapse
Affiliation(s)
- Shukui Qin
- Cancer Center of Jinling Hospital, Nanjing, China
| | - Jin Li
- Shanghai East Hospital, Shanghai, China
| | - Yuxian Bai
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Zishu Wang
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhendong Chen
- The Second Hospital of Anhui Medical University, Hefei, China
| | - Ruihua Xu
- Cancer Center of Sun Yat-sen University, Guangzhou, China
| | - Jianming Xu
- The Fifth Medical Center, General Hospital of People's Liberation Army, Beijing, China
| | - Hongmei Zhang
- Xijing Hospital, Air Force Medical University of PLA, Xi'an, China
| | - Jia Chen
- Jiangsu Cancer Hospital, Nanjing, China
| | - Ying Yuan
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianshu Liu
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lin Yang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haijun Zhong
- Zhejiang Cancer Hospital, Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, China
| | - Donghui Chen
- The First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Shen
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Chunyi Hao
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Deliang Fu
- Huashan Hospital, Fudan University, Shanghai, China
| | | | - Jianwei Yang
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Qiong Wang
- Jiangyin People's Hospital, Jiangyin, China
| | - Baoli Qin
- Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Hongming Pan
- Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Zhang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xianhong Bai
- Biotech Pharmaceutical Ltd, Corp, Beijing, China
| | - Qingshan Zheng
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
640
|
Greendyk JD, Allen WE, Alexander HR, Beninato T, Eskander MF, Grandhi MS, Kennedy TJ, Langan RC, Maggi JC, De S, Court CM, Ecker BL. Association between SMAD4 Mutations and GATA6 Expression in Paired Pancreatic Ductal Adenocarcinoma Tumor Specimens: Data from Two Independent Molecularly-Characterized Cohorts. Biomedicines 2023; 11:3058. [PMID: 38002058 PMCID: PMC10669842 DOI: 10.3390/biomedicines11113058] [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: 10/07/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Several molecular biomarkers have been identified to guide induction treatment selection for localized pancreatic ductal adenocarcinoma (PDAC). SMAD4 alterations and low GATA6 expression/modified "Moffitt" basal-like phenotype have each been associated with inferior survival uniquely for patients receiving 5-FU-based therapies. SMAD4 may directly regulate the expression of GATA6 in PDAC, pointing to a common predictive biomarker. To evaluate the relationship between SMAD4 mutations and GATA6 expression in human PDAC tumors, patients with paired SMAD4 mutation and GATA6 mRNA expression data in the TCGA and CPTAC were identified. In 321 patients (TCGA: n = 180; CPTAC: n = 141), the rate of SMAD4 alterations was 26.8%. The rate of SMAD4 alteration did not vary per tertile of normalized GATA6 expression (TCGA: p = 0.928; CPTAC: p = 0.828). In the TCGA, SMAD4 alterations and the basal-like phenotype were each associated with worse survival (log rank p = 0.077 and p = 0.080, respectively), but their combined presence did not identify a subset with uniquely inferior survival (p = 0.943). In the CPTAC, the basal-like phenotype was associated with significantly worse survival (p < 0.001), but the prognostic value was not influenced by the combined presence of SMAD4 alterations (p = 0.960). SMAD4 alterations were not associated with poor clinico-pathological features such as poor tumor grade, advanced tumor stage, positive lymphovascular invasion (LVI), or positive perineural invasion (PNI), compared with SMAD4-wildtype. Given that SMAD4 mutations were not associated with GATA6 expression or Moffitt subtype in two independent molecularly characterized PDAC cohorts, distinct biomarker-defined clinical trials are necessary.
Collapse
Affiliation(s)
- Joshua D. Greendyk
- Rutgers New Jersey Medical School, Rutgers Health, Newark, NJ 07103, USA; (J.D.G.); (W.E.A.)
| | - William E. Allen
- Rutgers New Jersey Medical School, Rutgers Health, Newark, NJ 07103, USA; (J.D.G.); (W.E.A.)
| | - H. Richard Alexander
- Rutgers Cancer Institute of New Jersey, Rutgers Health, New Brunswick, NJ 08901, USA; (H.R.A.); (T.B.); (M.F.E.); (M.S.G.); (T.J.K.); (R.C.L.); (J.C.M.); (S.D.)
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Toni Beninato
- Rutgers Cancer Institute of New Jersey, Rutgers Health, New Brunswick, NJ 08901, USA; (H.R.A.); (T.B.); (M.F.E.); (M.S.G.); (T.J.K.); (R.C.L.); (J.C.M.); (S.D.)
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Mariam F. Eskander
- Rutgers Cancer Institute of New Jersey, Rutgers Health, New Brunswick, NJ 08901, USA; (H.R.A.); (T.B.); (M.F.E.); (M.S.G.); (T.J.K.); (R.C.L.); (J.C.M.); (S.D.)
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Miral S. Grandhi
- Rutgers Cancer Institute of New Jersey, Rutgers Health, New Brunswick, NJ 08901, USA; (H.R.A.); (T.B.); (M.F.E.); (M.S.G.); (T.J.K.); (R.C.L.); (J.C.M.); (S.D.)
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Timothy J. Kennedy
- Rutgers Cancer Institute of New Jersey, Rutgers Health, New Brunswick, NJ 08901, USA; (H.R.A.); (T.B.); (M.F.E.); (M.S.G.); (T.J.K.); (R.C.L.); (J.C.M.); (S.D.)
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Russell C. Langan
- Rutgers Cancer Institute of New Jersey, Rutgers Health, New Brunswick, NJ 08901, USA; (H.R.A.); (T.B.); (M.F.E.); (M.S.G.); (T.J.K.); (R.C.L.); (J.C.M.); (S.D.)
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
- Cooperman Barnabas Medical Center, Livingston, NJ 07039, USA
| | - Jason C. Maggi
- Rutgers Cancer Institute of New Jersey, Rutgers Health, New Brunswick, NJ 08901, USA; (H.R.A.); (T.B.); (M.F.E.); (M.S.G.); (T.J.K.); (R.C.L.); (J.C.M.); (S.D.)
- Cooperman Barnabas Medical Center, Livingston, NJ 07039, USA
| | - Subhajyoti De
- Rutgers Cancer Institute of New Jersey, Rutgers Health, New Brunswick, NJ 08901, USA; (H.R.A.); (T.B.); (M.F.E.); (M.S.G.); (T.J.K.); (R.C.L.); (J.C.M.); (S.D.)
| | - Colin M. Court
- Department of Surgical Oncology, University of Texas San Antonio, San Antonio, TX 78249, USA;
| | - Brett L. Ecker
- Rutgers Cancer Institute of New Jersey, Rutgers Health, New Brunswick, NJ 08901, USA; (H.R.A.); (T.B.); (M.F.E.); (M.S.G.); (T.J.K.); (R.C.L.); (J.C.M.); (S.D.)
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
- Cooperman Barnabas Medical Center, Livingston, NJ 07039, USA
| |
Collapse
|
641
|
Singh H, Keller RB, Kapner KS, Dilly J, Raghavan S, Yuan C, Cohen EF, Tolstorukov M, Andrews E, Brais LK, Da Silva A, Perez K, Rubinson DA, Surana R, Giannakis M, Ng K, Clancy TE, Yurgelun MB, Schletchter B, Clark JW, Shapiro GI, Rosenthal MH, Hornick JL, Nardi V, Li YY, Gupta H, Cherniack AD, Meyerson M, Cleary JM, Nowak JA, Wolpin BM, Aguirre AJ. Oncogenic Drivers and Therapeutic Vulnerabilities in KRAS Wild-Type Pancreatic Cancer. Clin Cancer Res 2023; 29:4627-4643. [PMID: 37463056 PMCID: PMC10795103 DOI: 10.1158/1078-0432.ccr-22-3930] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/17/2023] [Accepted: 07/14/2023] [Indexed: 07/20/2023]
Abstract
PURPOSE Approximately 8% to 10% of pancreatic ductal adenocarcinomas (PDAC) do not harbor mutations in KRAS. Understanding the unique molecular and clinical features of this subset of pancreatic cancer is important to guide patient stratification for clinical trials of molecularly targeted agents. EXPERIMENTAL DESIGN We analyzed a single-institution cohort of 795 exocrine pancreatic cancer cases (including 785 PDAC cases) with a targeted multigene sequencing panel and identified 73 patients (9.2%) with KRAS wild-type (WT) pancreatic cancer. RESULTS Overall, 43.8% (32/73) of KRAS WT cases had evidence of an alternative driver of the MAPK pathway, including BRAF mutations and in-frame deletions and receptor tyrosine kinase fusions. Conversely, 56.2% of cases did not harbor a clear MAPK driver alteration, but 29.3% of these MAPK-negative KRAS WT cases (12/41) demonstrated activating alterations in other oncogenic drivers, such as GNAS, MYC, PIK3CA, and CTNNB1. We demonstrate potent efficacy of pan-RAF and MEK inhibition in patient-derived organoid models carrying BRAF in-frame deletions. Moreover, we demonstrate durable clinical benefit of targeted therapy in a patient harboring a KRAS WT tumor with a ROS1 fusion. Clinically, patients with KRAS WT tumors were significantly younger in age of onset (median age: 62.6 vs. 65.7 years; P = 0.037). SMAD4 mutations were associated with a particularly poor prognosis in KRAS WT cases. CONCLUSIONS This study defines the genomic underpinnings of KRAS WT pancreatic cancer and highlights potential therapeutic avenues for future investigation in molecularly directed clinical trials. See related commentary by Kato et al., p. 4527.
Collapse
Affiliation(s)
- Harshabad Singh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Rachel B. Keller
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Kevin S. Kapner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Julien Dilly
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Biological and biomedical sciences program, Harvard Medical School, Boston, MA
- The Broad Institute of Harvard and MIT, Cambridge, MA
| | - Srivatsan Raghavan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- The Broad Institute of Harvard and MIT, Cambridge, MA
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Elizabeth F. Cohen
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA
| | - Michael Tolstorukov
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA
| | - Elizabeth Andrews
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Lauren K. Brais
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Annacarolina Da Silva
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Pathology, Weill Cornell Medical College, New York, NY
| | - Kimberly Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Douglas A. Rubinson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Rishi Surana
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Thomas E. Clancy
- Harvard Medical School, Boston, MA
- Division of Surgical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA
| | - Matthew B. Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Benjamin Schletchter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Jeffrey W. Clark
- Harvard Medical School, Boston, MA
- Massachusetts General Hospital Cancer Center, Boston, MA
| | - Geoffrey I. Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Michael H. Rosenthal
- Department of Radiology, Dana-Farber Cancer Institute, Boston, MA
- Department of Radiology, Brigham and Women’s Hospital, Boston, MA
| | - Jason L. Hornick
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Yvonne Y. Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- The Broad Institute of Harvard and MIT, Cambridge, MA
| | - Hersh Gupta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- The Broad Institute of Harvard and MIT, Cambridge, MA
| | - Andrew D. Cherniack
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- The Broad Institute of Harvard and MIT, Cambridge, MA
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- The Broad Institute of Harvard and MIT, Cambridge, MA
| | - James M. Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Jonathan A. Nowak
- Harvard Medical School, Boston, MA
- Department of Radiology, Brigham and Women’s Hospital, Boston, MA
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Andrew J. Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- The Broad Institute of Harvard and MIT, Cambridge, MA
| |
Collapse
|
642
|
Chen Z, He Y, Ding C, Chen J, Gu Y, Xiao M, Li Q. Safety and Efficacy Analysis of PD-1 Inhibitors in Combination with Gemcitabine Plus Nab-Paclitaxel for Advanced Pancreatic Cancer: A Real-World, Single-Center Study. Onco Targets Ther 2023; 16:923-935. [PMID: 37965584 PMCID: PMC10642393 DOI: 10.2147/ott.s427942] [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: 06/28/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023] Open
Abstract
Background Pancreatic cancer is a deadly disease with a low five years survival rate, and chemotherapy remains the standard treatment for advanced cases. However, the efficacy of chemotherapy alone is limited, and there is a need for new treatment options. Recently, immune checkpoint inhibitors (ICIs), particularly programmed death-1 (PD-1) inhibitors, have shown promising results in various cancers, including pancreatic cancer. In this study, we explore the safety and efficacy of PD-1 inhibitors in combination with chemotherapy for advanced pancreatic cancer. Materials and Methods A retrospective analysis was conducted on clinical data from 27 patients with advanced pancreatic cancer who were administered a combination of anti-PD-1 antibody and gemcitabine plus nab-paclitaxel (GnP) regimen. The study evaluated the safety of the treatment as well as the objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). Results In this study, treatment with a combination of anti-PD-1 antibody and GnP regimen for pancreatic cancer resulted in partial response (PR) for 10 out of 27 (37.04%) patients, stable disease (SD) for 10 (37.04%) patients, and progressive disease (PD) for 7 (25.92%) patients. The study found that the median OS (mOS) for these patients was 16.4 months [standard error (SE) = 1.117, 95% confidence interval (CI) 14.211-18.589], while the median PFS (mPFS) was 6.4 months (SE = 1.217, 95% CI 3.981-8.752). Subgroup analysis revealed that pancreatic cancer patients' Eastern Cooperative Oncology Group (ECOG) performance status (PS) (0 vs 1) and treatment cycles (≤6 cycles vs >6 cycles) significantly affected OS and PFS. Patients experienced mostly grade 1-2 adverse events (AEs), which were relieved through clinical treatment. Conclusion The combination of GnP with anti-PD-1 antibodies shows promise as a potential treatment option for advanced pancreatic cancer.
Collapse
Affiliation(s)
- Zhitao Chen
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| | - Yahui He
- School of Medicine, Zhejiang Chinese Medical University Zhejiang Shuren College, Hangzhou, 310003, People’s Republic of China
| | - Chenchen Ding
- Child and Adolescent Psychology, Affiliated Mental Health Centre & Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310013, People’s Republic of China
| | - Jun Chen
- School of Medicine, Zhejiang Chinese Medical University Zhejiang Shuren College, Hangzhou, 310003, People’s Republic of China
| | - Yangjun Gu
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| | - Min Xiao
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| | - Qiyong Li
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| |
Collapse
|
643
|
Fukahori M, Okabe Y, Shimokawa M, Otsuka T, Koga F, Ueda Y, Nakazawa J, Komori A, Otsu S, Arima S, Makiyama A, Taguchi H, Honda T, Ushijima T, Miwa K, Shibuki T, Nio K, Ide Y, Ureshino N, Mizuta T, Mitsugi K, Shirakawa T. Efficacy of second-line chemotherapy after treatment with gemcitabine plus nab-paclitaxel or FOLFIRINOX in patients with metastatic pancreatic cancer. Sci Rep 2023; 13:19399. [PMID: 37938630 PMCID: PMC10632451 DOI: 10.1038/s41598-023-46924-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 11/07/2023] [Indexed: 11/09/2023] Open
Abstract
First-line chemotherapy for patients with metastatic pancreatic cancer (MPC) includes gemcitabine plus nab-paclitaxel (GnP) and FOLFIRINOX (FFX). However, the efficacy of second-line chemotherapy and the role of combination chemotherapy in clinical practice is still unknown. Data was gathered from 14 hospitals in the Kyushu area of Japan from December 2013 to March 2017. The median overall survival (mOS) from second-line treatment was contrasted between patients who received second-line chemotherapy (CT group) and those who received the best supportive care (BSC group). Furthermore, the mOS of combination chemotherapy was compared to mono chemotherapy in the CT group. To control possible bias in the selection of treatment, we performed a propensity score-adjusted analysis. A total of 255 patients received GnP or FFX as first-line chemotherapy. There were 156 in the CT group and 77 in the BSC group of these. The CT group had a significantly longer mOS than the BSC group (5.2 vs. 2.6 months; adjusted hazard ratio (HR) 0.38; 95% CI 0.27-0.54). In the CT group, 89 patients received combination chemotherapy while 67 received mono chemotherapy. The mOS did not differ significantly between the combination and mono chemotherapy groups (5.5 vs. 4.8 months; adjusted HR 0.88; 95% CI 0.58-1.33). Among patients with MPC receiving second-line treatment, the CT group had a significantly longer mOS than the BSC group, but combination chemotherapy conferred no improvement in survival compared to mono chemotherapy.
Collapse
Affiliation(s)
- Masaru Fukahori
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume-Shi, Fukuoka, 830-0011, Japan
- Kyoto Innovation Center for Next Generation Clinical Trials and iPS Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yoshinobu Okabe
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume-Shi, Fukuoka, 830-0011, Japan
| | - Mototsugu Shimokawa
- Clinical Research Institute, National Kyushu Cancer Center, 3-1-1 Notame, Minami-Ku, Fukuoka-Shi, Fukuoka, 811-1395, Japan
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube-Shi, Yamaguchi, 755-8505, Japan
| | - Taiga Otsuka
- Department of Medical Oncology, Saga Medical Center Koseikan, 400 Kase-Machi, Saga-Shi, Saga, 840-8571, Japan.
- Department of Internal Medicine, Minato Medical Clinic, 3-11-3 Nagahama, Chuo-Ku, Fukuoka-Shi, Fukuoka, 810-0072, Japan.
| | - Futa Koga
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, 400 Kase-Machi, Saga-Shi, Saga, 840-8571, Japan
| | - Yujiro Ueda
- Department of Hematology and Oncology, Japanese Red Cross Kumamoto Hospital, 2-1-1 Nagamine-Minami, Higashi-Ku, Kumamoto-Shi, Kumamoto, 861-8520, Japan
| | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-Cho, Kagoshima-Shi, Kagoshima, 890-8760, Japan
| | - Azusa Komori
- Department of Medical Oncology and Hematology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-Machi, Yufu-Shi, Oita, 879-5593, Japan
| | - Satoshi Otsu
- Department of Medical Oncology and Hematology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-Machi, Yufu-Shi, Oita, 879-5593, Japan
| | - Shiho Arima
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima-Shi, Kagoshima, 890-8520, Japan
| | - Akitaka Makiyama
- Department of Hematology Oncology, Japan Community Healthcare Organization Kyushu Hospital, 1-8-1 Kishinoura, Yahatanishi-Ku, Kitakyushu-Shi, Fukuoka, 806-8501, Japan
- Cancer Center, Gifu University Hospital, 1-1 Yanagido, Gifu-Shi, Gifu, 501-1194, Japan
| | - Hiroki Taguchi
- Department of Gastroenterology, Saiseikai Sendai Hospital, 2-46 Harada-Cho, Satsumasendai-Shi, Kagoshima, 895-0074, Japan
- Department of Gastroenterology, Imamura General Hospital, 11-23 Kamoike-Shinmachi, Kagoshima-Shi, Kagoshima, 890-0064, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki-Shi, Nagasaki, 852-8501, Japan
| | - Tomoyuki Ushijima
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume-Shi, Fukuoka, 830-0011, Japan
| | - Keisuke Miwa
- Multidisciplinary Treatment Cancer Center, Kurume University Hospital, 67 Asahi-Machi, Kurume-Shi, Fukuoka, 830-0011, Japan
| | - Taro Shibuki
- Department of Internal Medicine, Imari Arita Kyoritsu Hospital, 860 Ninose-Ko, Arita-Cho, Nishi-Matsuura-Gun, Saga, 849-4193, Japan
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanohara, Kashiwa-Shi, Chiba, 277-8577, Japan
| | - Kenta Nio
- Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-Cho, Sasebo-Shi, Nagasaki, 857-8575, Japan
- Department of Medical Oncology, Hamanomachi Hospital, 3-3-1 Nagahama, Chuo-Ku, Fukuoka-Shi, Fukuoka, 810-8539, Japan
| | - Yasushi Ide
- Department of Internal Medicine, Karatsu Red Cross Hospital, 2430 Watada, Karatsu-Shi, Saga, 847-8588, Japan
| | - Norio Ureshino
- Department of Medical Oncology, Saga Medical Center Koseikan, 400 Kase-Machi, Saga-Shi, Saga, 840-8571, Japan
- Department of Medical Oncology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-Shi, Chiba, 292-8535, Japan
| | - Toshihiko Mizuta
- Department of Internal Medicine, Imari Arita Kyoritsu Hospital, 860 Ninose-Ko, Arita-Cho, Nishi-Matsuura-Gun, Saga, 849-4193, Japan
- Department of Internal Medicine, Fujikawa Hospital, 1-2-6 Matsubara, Saga-Shi, Saga, 840-0831, Japan
| | - Kenji Mitsugi
- Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-Cho, Sasebo-Shi, Nagasaki, 857-8575, Japan
- Department of Medical Oncology, Hamanomachi Hospital, 3-3-1 Nagahama, Chuo-Ku, Fukuoka-Shi, Fukuoka, 810-8539, Japan
| | - Tsuyoshi Shirakawa
- Department of Medical Oncology, Fukuoka Wajiro Hospital, 2-2-75 Wajirogaoka, Higashi-Ku, Fukuoka-Shi, Fukuoka, 811-0213, Japan
- Department of Internal Medicine, Karatsu Higashi-Matsuura Medical Association Center, 2566-11 Chiyoda-machi, Karatsu-Shi, Saga, 847-0041, Japan
| |
Collapse
|
644
|
Hau SO, Svensson M, Petersson A, Eberhard J, Jirström K. Trajectories of immune-related serum proteins and quality of life in patients with pancreatic and other periampullary cancer: the CHAMP study. BMC Cancer 2023; 23:1074. [PMID: 37936126 PMCID: PMC10629201 DOI: 10.1186/s12885-023-11562-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND There is still a profound lack of efficient therapeutic strategies against pancreatic and other periampullary adenocarcinoma. Surgery is seldom possible, leaving palliative chemotherapy the only option for most patients. Chemotherapy treatment is however often accompanied by serious side-effects, and the identification of biomarkers for early prediction of disease and treatment-associated symptoms could help alleviate patient suffering. This study investigated the dynamic interrelationship between immune-related serum proteins, routine biomarkers, and health-related quality of life (HRQoL) factors during chemotherapy treatment of patients enrolled in the prospective, observational study Chemotherapy, Host response And Molecular dynamics in Periampullary cancer (CHAMP). METHODS Proximity extension assay was applied to analyse 92 immune-associated proteins in longitudinal serum samples from 75 patients, 18 treated with curative and 57 with palliative intent. HRQoL data were available from all patients at baseline (BL), from 41 patients at three months, and from 23 patients at six months. Information on routine laboratory parameters albumin, CA19-9, CEA and CRP were collected from medical charts. RESULTS In total nine proteins; chemokine (C-C motif) ligand 23 (CCL23), cluster of differentiation 4 (CD4), cluster of differentiation 28 (CD28), decorin (DCN), galectin-1 (Gal-1), granzyme B (GZMB), granzyme H (GZMH), matrix metallopeptidase 7 (MMP7), and monocyte chemotactic protein-1 (MCP-1) were strongly correlated (Spearman's Rho ≤ -0.6 or ≥ 0.6) with either cognitive functioning (DCN), emotional functioning (DCN, MCP-1), dyspnoea (CD28, GZMB, GZMH) or insomnia (CCL23, CD4, Gal-1, MMP7) during treatment. Associations between routine laboratory parameters (CA 19-9, CA-125, CRP, CEA and albumin) and HRQoL factors were overall weaker. None of the investigated proteins were associated with pain. CONCLUSIONS This is, to our knowledge, the first study exploring associations between serum biomarkers and HRQoL in patients with pancreatic or other periampullary cancer, and some findings merit further validation. The associations of DCN and MCP-1with impaired cognitive and/or emotional functioning are of particular interest, given their established link to various neurodegenerative conditions. Chemotherapy is known to cause persistent cognitive dysfunction with effects on memory and executive function, referred to as "chemo brain". It would therefore be of great value to identify biomarkers for early detection and management of this debilitating condition. TRIAL REGISTRATION Clinical Trial Registration: NCT03724994.
Collapse
Affiliation(s)
- Sofie Olsson Hau
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - Maja Svensson
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Alexandra Petersson
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jakob Eberhard
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Karin Jirström
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
645
|
Park MN. Therapeutic Strategies for Pancreatic-Cancer-Related Type 2 Diabetes Centered around Natural Products. Int J Mol Sci 2023; 24:15906. [PMID: 37958889 PMCID: PMC10648679 DOI: 10.3390/ijms242115906] [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: 08/25/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a highly malignant neoplasm, is classified as one of the most severe and devastating types of cancer. PDAC is a notable malignancy that exhibits a discouraging prognosis and a rising occurrence. The interplay between diabetes and pancreatic cancer exhibits a reciprocal causation. The identified metabolic disorder has been observed to possess noteworthy consequences on health outcomes, resulting in elevated rates of morbidity. The principal mechanisms involve the suppression of the immune system, the activation of pancreatic stellate cells (PSCs), and the onset of systemic metabolic disease caused by dysfunction of the islets. From this point forward, it is important to recognize that pancreatic-cancer-related diabetes (PCRD) has the ability to increase the likelihood of developing pancreatic cancer. This highlights the complex relationship that exists between these two physiological states. Therefore, we investigated into the complex domain of PSCs, elucidating their intricate signaling pathways and the profound influence of chemokines on their behavior and final outcome. In order to surmount the obstacle of drug resistance and eliminate PDAC, researchers have undertaken extensive efforts to explore and cultivate novel natural compounds of the next generation. Additional investigation is necessary in order to comprehensively comprehend the effect of PCRD-mediated apoptosis on the progression and onset of PDAC through the utilization of natural compounds. This study aims to examine the potential anticancer properties of natural compounds in individuals with diabetes who are undergoing chemotherapy, targeted therapy, or immunotherapy. It is anticipated that these compounds will exhibit increased potency and possess enhanced pharmacological benefits. According to our research findings, it is indicated that naturally derived chemical compounds hold potential in the development of PDAC therapies that are both safe and efficacious.
Collapse
Affiliation(s)
- Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Republic of Korea
| |
Collapse
|
646
|
Hu X, Hu D, Fu B, Li H, Ren G, Liu H, Song J, Kang X, Wang X, Pang H, Liu C, Zhang J, Wang Y. Survival benefit of local consolidative therapy for patients with single-organ metastatic pancreatic cancer: a propensity score-matched cross-sectional study based on 17 registries. Front Endocrinol (Lausanne) 2023; 14:1225979. [PMID: 38027134 PMCID: PMC10652880 DOI: 10.3389/fendo.2023.1225979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Background The continuous exploration of oligometastatic disease has led to the remarkable achievements of local consolidative therapy (LCT) and favorable outcomes for this disease. Thus, this study investigated the potential benefits of LCT in patients with single-organ metastatic pancreatic ductal adenocarcinoma (PDAC). Methods Patients with single-organ metastatic PDAC diagnosed between 2010 - 2019 were identified from the Surveillance, Epidemiology, and End Results (SEER) database. Propensity score matching (PSM) was performed to minimize selection bias. Factors affecting survival were assessed by Cox regression analysis and Kaplan-Meier estimates. Results A total of 12900 patients were identified from the database, including 635 patients who received chemotherapy combined with LCT with a 1:1 PSM with patients who received only chemotherapy. Patients with single-organ metastatic PDAC who received chemotherapy in combination with LCT demonstrated extended median overall survival (OS) by approximately 57%, more than those who underwent chemotherapy alone (11 vs. 7 months, p < 0.001). Furthermore, the multivariate Cox regression analysis revealed that patients that received LCT, younger age (< 65 years), smaller tumor size (< 50 mm), and lung metastasis (reference: liver) were favorable prognostic factors for patients with single-organ metastatic PDAC. Conclusion The OS of patients with single-organ metastatic pancreatic cancer who received LCT may be prolonged compared to those who received only chemotherapy. Nevertheless, additional prospective randomized clinical trials are required to support these findings.
Collapse
Affiliation(s)
- Xiaolong Hu
- Department of Radiation Oncology, Beijing Geriatric Hospital, Beijing, China
| | - Dan Hu
- Outpatient Department of Feng Tai District No.4 Retired Cadres Retreat Center, Army PLA, Beijing, China
| | - Bowen Fu
- Department of Radiation Oncology, Air Force Medical Center PLA, Beijing, China
| | - Hongqi Li
- Department of Radiation Oncology, Air Force Medical Center PLA, Beijing, China
| | - Gang Ren
- Department of Radiation Oncology, Peking University Shou Gang Hospital, Beijing, China
| | - Hefei Liu
- Center for Ion Medicine, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
| | - Jiazhao Song
- Department of Radiation Oncology, Air Force Medical Center PLA, Beijing, China
| | - Xiaoli Kang
- Department of Radiation Oncology, Air Force Medical Center PLA, Beijing, China
| | - Xuan Wang
- Department of Radiation Oncology, Air Force Medical Center PLA, Beijing, China
| | - Haifeng Pang
- Department of Radiation Oncology, Air Force Medical Center PLA, Beijing, China
| | - Chen Liu
- Department of Radiation Oncology, Air Force Medical Center PLA, Beijing, China
| | - Jianchun Zhang
- Department of Radiation Oncology, Beijing Geriatric Hospital, Beijing, China
| | - Yingjie Wang
- Department of Radiation Oncology, Air Force Medical Center PLA, Beijing, China
| |
Collapse
|
647
|
Conroy T, Pfeiffer P, Vilgrain V, Lamarca A, Seufferlein T, O'Reilly EM, Hackert T, Golan T, Prager G, Haustermans K, Vogel A, Ducreux M. Pancreatic cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2023; 34:987-1002. [PMID: 37678671 DOI: 10.1016/j.annonc.2023.08.009] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Affiliation(s)
- T Conroy
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandoeuvre-lès-Nancy; APEMAC, équipe MICS, Université de Lorraine, Nancy, France
| | - P Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - V Vilgrain
- Centre de Recherche sur l'Inflammation U 1149, Université Paris Cité, Paris; Department of Radiology, Beaujon Hospital, APHP Nord, Clichy, France
| | - A Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - T Seufferlein
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - E M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - T Hackert
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - T Golan
- Gastrointestinal Unit, Oncology Institute, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - G Prager
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - K Haustermans
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - A Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - M Ducreux
- Université Paris-Saclay, Gustave Roussy, Inserm Unité Dynamique des Cellules Tumorales, Villejuif, France
| |
Collapse
|
648
|
Evrard C, Ingrand P, Rochelle T, Martel M, Tachon G, Flores N, Randrian V, Ferru A, Haineaux PA, Goujon JM, Karayan-Tapon L, Tougeron D. Circulating tumor DNA in unresectable pancreatic cancer is a strong predictor of first-line treatment efficacy: The KRASCIPANC prospective study. Dig Liver Dis 2023; 55:1562-1572. [PMID: 37308396 DOI: 10.1016/j.dld.2023.03.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/08/2023] [Accepted: 03/27/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND There is no robust predictor of response to chemotherapy (CT) in unresectable pancreatic adenocarcinomas (UPA). The objective of the KRASCIPANC study was to analyze the kinetics of cell-free DNA (cfDNA)/circulating tumor DNA (ctDNA) as a predictor of response to CT in UPA. METHODS Blood samples were collected just before first CT and at day 28. The primary endpoint was the kinetics of KRAS-mutated ctDNA by digital droplet PCR between D0 and D28 as a predictor of progression-free survival (PFS). RESULTS We analyzed 65 patients with a KRAS-mutated tumor. A high level of cfDNA and KRAS-mutated ctDNA at D0, as well as the presence of KRAS-mutated ctDNA at D28, were strongly associated with lower centralized disease control rate (cDCR), shorter cPFS and OS in multivariate analysis. A score combining cfDNA level at diagnosis ≥ or <30 ng/mL and presence or not of KRAS-mutated ctDNA at D28 was an optimal predictor of cDCR (OR=30.7, IC95% 4.31-218 P=.001), PFS (HR=6.79, IC95% 2.76-16.7, P<.001) and OS (HR=9.98, IC95% 4.14-24.1, P<.001). CONCLUSION A combined score using cfDNA level at diagnosis and KRAS-mutated ctDNA at D28 is strongly associated with patient survival/response to chemotherapy in UPA. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04560270.
Collapse
Affiliation(s)
- Camille Evrard
- Medical Oncology Department, Poitiers University Hospital, Poitiers 86000, France; ProDicET, UR 24144, University of Poitiers, Poitiers 86000, France.
| | - Pierre Ingrand
- Department of Statistics, Faculty of Medicine, University of Poitiers, Poitiers 86000, France
| | - Tristan Rochelle
- Cancer Biology Department, Poitiers University Hospital, Poitiers 86000, France
| | - Marine Martel
- Cancer Biology Department, Poitiers University Hospital, Poitiers 86000, France
| | - Gaëlle Tachon
- Cancer Biology Department, Poitiers University Hospital, Poitiers 86000, France; Cancer Biology Department, Centre Léon Bérard, Lyon 69000, France
| | - Nicolas Flores
- Department of Imaging, University Hospital of Poitiers, Poitiers 86000, France
| | - Violaine Randrian
- ProDicET, UR 24144, University of Poitiers, Poitiers 86000, France; Hepato-Gastroenterology Department, Poitiers University Hospital, Poitiers, France
| | - Aurélie Ferru
- Medical Oncology Department, Poitiers University Hospital, Poitiers 86000, France
| | - Paul-Arthur Haineaux
- Hepato-Gastroenterology Department, Poitiers University Hospital, Poitiers, France; Hepato-Gastroenterology Department, Poitiers University Hospital, Châtellerault Hospital, Poitiers 86106, France
| | - Jean-Michel Goujon
- Department of Pathology, Poitiers University Hospital, Poitiers 86000, France
| | - Lucie Karayan-Tapon
- ProDicET, UR 24144, University of Poitiers, Poitiers 86000, France; Cancer Biology Department, Poitiers University Hospital, Poitiers 86000, France
| | - David Tougeron
- ProDicET, UR 24144, University of Poitiers, Poitiers 86000, France; Hepato-Gastroenterology Department, Poitiers University Hospital, Poitiers, France.
| |
Collapse
|
649
|
Ruiz CF, Garcia C, Jacox JB, Lawres L, Muzumdar MD. Decoding the obesity-cancer connection: lessons from preclinical models of pancreatic adenocarcinoma. Life Sci Alliance 2023; 6:e202302228. [PMID: 37648285 PMCID: PMC10474221 DOI: 10.26508/lsa.202302228] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
Obesity is a metabolic state of energy excess and a risk factor for over a dozen cancer types. Because of the rising worldwide prevalence of obesity, decoding the mechanisms by which obesity promotes tumor initiation and early progression is a societal imperative and could broadly impact human health. Here, we review results from preclinical models that link obesity to cancer, using pancreatic adenocarcinoma as a paradigmatic example. We discuss how obesity drives cancer development by reprogramming the pretumor or tumor cell and its micro- and macro-environments. Specifically, we describe evidence for (1) altered cellular metabolism, (2) hormone dysregulation, (3) inflammation, and (4) microbial dysbiosis in obesity-driven pancreatic tumorigenesis, denoting variables that confound interpretation of these studies, and highlight remaining gaps in knowledge. Recent advances in preclinical modeling and emerging unbiased analytic approaches will aid in further unraveling the complex link between obesity and cancer, informing novel strategies for prevention, interception, and therapy in pancreatic adenocarcinoma and other obesity-associated cancers.
Collapse
Affiliation(s)
- Christian F Ruiz
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Cathy Garcia
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Jeremy B Jacox
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
- Department of Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, CT, USA
| | - Lauren Lawres
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Mandar D Muzumdar
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
- Department of Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale University, New Haven, CT, USA
| |
Collapse
|
650
|
De Monte L, Clemente F, Ruggiero E, Pini R, Ceraolo MG, Schiavo Lena M, Balestrieri C, Lazarevic D, Belfiori G, Crippa S, Balzano G, Falconi M, Doglioni C, Bonini C, Reni M, Protti MP. Pro-tumor Tfh2 cells induce detrimental IgG4 production and PGE 2-dependent IgE inhibition in pancreatic cancer. EBioMedicine 2023; 97:104819. [PMID: 37776595 PMCID: PMC10542011 DOI: 10.1016/j.ebiom.2023.104819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis and it is characterized by predominant pro-tumor Th2-type inflammation. T follicular helper (Tfh) cells are relevant immunoregulators in cancer, and often correlate with better survival. How the Th2-skewed microenvironment in PDAC modulates the differentiation of Tfh cells and their immunoregulatory function is unknown. METHODS We carried out high-dimensional flow cytometry and T-cell receptor- and RNA-sequencing, as well as bioinformatics, immunohistochemistry and in vitro mechanistic studies. FINDINGS We identified Tfh1-, Tfh2-, and Tfh17-like cell clusters in the blood, tumors and tumor-draining lymph-nodes (TDLNs) of chemo-naïve PDAC patients and showed that high percentages of Tfh2 cells within the tumor tissue and TDLNs correlated with reduced patient survival. Moreover, only Tfh2 cells were highly activated and were reduced in frequency in patients who responded to neoadjuvant chemotherapy. RNA-sequencing analysis of immunoglobulin expression showed that tumor and TDLN samples expressed all immunoglobulin (IGH) isotypes apart from IGHE. Consistent with these findings, Tfh2 cells differentiated in vitro by tumor microenvironment-conditioned dendritic cells promoted the production of anti-inflammatory IgG4 antibodies by co-cultured B cells, dependent on IL-13. Moreover, unexpectedly, Tfh2 cells inhibited the secretion of pro-inflammatory IgE, dependent on prostaglandin E2. INTERPRETATION Our results indicate that in PDAC, highly activated pro-tumor Tfh2 favor anti-inflammatory IgG4 production, while inhibit pro-inflammatory IgE. Thus, targeting the circuits that drive Tfh2 cells, in combination with chemotherapy, may re-establish beneficial anti-tumor Tfh-B cell interactions and facilitate more effective treatment. FUNDING Research grants from the Italian Association for Cancer Research (AIRC) IG-19119 to MPP and the AIRC Special Program in Metastatic disease: the key unmet need in oncology, 5 per Mille no. 22737 to CB, MF, CD, MR and MPP; the ERA-NET EuroNanoMed III (a collaborative european grant financed by the Italian Ministry of Health, Italy) project PANIPAC (JTC2018/041) to MPP; the Fondazione Valsecchi to SC.
Collapse
Affiliation(s)
- Lucia De Monte
- Tumor Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy; Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Clemente
- Tumor Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy; Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eliana Ruggiero
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Pini
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Grazia Ceraolo
- Tumor Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy; Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Chiara Balestrieri
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Dejan Lazarevic
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Belfiori
- Pancreatic Surgery Unit and Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Crippa
- Pancreatic Surgery Unit and Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Gianpaolo Balzano
- Pancreatic Surgery Unit and Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit and Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Claudio Doglioni
- Pathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Chiara Bonini
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Michele Reni
- Vita-Salute San Raffaele University, Milan, Italy; Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Pia Protti
- Tumor Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy; Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|