401
|
De Santis MC, Bockorny B, Hirsch E, Cappello P, Martini M. Exploiting pancreatic cancer metabolism: challenges and opportunities. Trends Mol Med 2024; 30:592-604. [PMID: 38604929 DOI: 10.1016/j.molmed.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 04/13/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive form of pancreatic cancer, known for its challenging diagnosis and limited treatment options. The focus on metabolic reprogramming as a key factor in tumor initiation, progression, and therapy resistance has gained prominence. In this review we focus on the impact of metabolic changes on the interplay among stromal, immune, and tumor cells, as glutamine and branched-chain amino acids (BCAAs) emerge as pivotal players in modulating immune cell functions and tumor growth. We also discuss ongoing clinical trials that explore metabolic modulation for PDAC, targeting mitochondrial metabolism, asparagine and glutamine addiction, and autophagy inhibition. Overcoming challenges in understanding nutrient effects on immune-stromal-tumor interactions holds promise for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Maria Chiara De Santis
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy.
| | - Bruno Bockorny
- BIDMC Department of Medicine, Harvard Medical School, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Miriam Martini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy.
| |
Collapse
|
402
|
Maino C, Cereda M, Franco PN, Boraschi P, Cannella R, Gianotti LV, Zamboni G, Vernuccio F, Ippolito D. Cross-sectional imaging after pancreatic surgery: The dialogue between the radiologist and the surgeon. Eur J Radiol Open 2024; 12:100544. [PMID: 38304573 PMCID: PMC10831502 DOI: 10.1016/j.ejro.2023.100544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/29/2023] [Accepted: 12/29/2023] [Indexed: 02/03/2024] Open
Abstract
Pancreatic surgery is nowadays considered one of the most complex surgical approaches and not unscathed from complications. After the surgical procedure, cross-sectional imaging is considered the non-invasive reference standard to detect early and late compilations, and consequently to address patients to the best management possible. Contras-enhanced computed tomography (CECT) should be considered the most important and useful imaging technique to evaluate the surgical site. Thanks to its speed, contrast, and spatial resolution, it can help reach the final diagnosis with high accuracy. On the other hand, magnetic resonance imaging (MRI) should be considered as a second-line imaging approach, especially for the evaluation of biliary findings and late complications. In both cases, the radiologist should be aware of protocols and what to look at, to create a robust dialogue with the surgeon and outline a fitted treatment for each patient.
Collapse
Affiliation(s)
- Cesare Maino
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, MB, Italy
| | - Marco Cereda
- Department of Surgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, MB, Italy
| | - Paolo Niccolò Franco
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, MB, Italy
| | - Piero Boraschi
- Radiology Unit, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy
| | - Roberto Cannella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Luca Vittorio Gianotti
- Department of Surgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, MB, Italy
- School of Medicine, Università Milano-Bicocca, Piazza dell’Ateneo Nuovo, 1, 20100 Milano, Italy
| | - Giulia Zamboni
- Institute of Radiology, Department of Diagnostics and Public Health, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Federica Vernuccio
- University Hospital of Padova, Institute of Radiology, 35128 Padova, Italy
| | - Davide Ippolito
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, MB, Italy
- School of Medicine, Università Milano-Bicocca, Piazza dell’Ateneo Nuovo, 1, 20100 Milano, Italy
| |
Collapse
|
403
|
Ito M, Okumura Y, Nio K, Baba E, Ozaki Y, Nishio H, Ichihara E, Miura Y, Endo M, Yano S, Maruyama D, Yoshinami T, Susumu N, Takekuma M, Motohashi T, Ochi N, Kubo T, Uchino K, Kimura T, Kamiyama Y, Nakao S, Tamura S, Nishimoto H, Kato Y, Sato A, Takano T, Tsuchihashi K. Effectiveness of G-CSF in chemotherapy for digestive system tumors: a systematic review of the Clinical Practice Guidelines for the Use of G-CSF 2022 delineated by the Japan Society of Clinical Oncology. Int J Clin Oncol 2024; 29:689-699. [PMID: 38578596 DOI: 10.1007/s10147-024-02502-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/04/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Granulocyte colony-stimulating factor (G-CSF) reportedly reduces the risk of neutropenia and subsequent infections caused by cancer chemotherapy. Although several guidelines recommend using G-CSF in primary prophylaxis according to the incidence rate of chemotherapy-induced febrile neutropenia (FN), the effectiveness of G-CSF in digestive system tumor chemotherapy remains unclear. To address these clinical questions, we conducted a systematic review as part of revising the Clinical Practice Guidelines for the Use of G-CSF 2022 published by the Japan Society of Clinical Oncology. METHODS This systematic review addressed two main clinical questions (CQ): CQ1: "Is primary prophylaxis with G-CSF effective in chemotherapy?", and CQ2: "Is increasing the intensity of chemotherapy with G-CSF effective?" We reviewed different types of digestive system tumors, including esophageal, gastric, pancreatic, biliary tract, colorectal, and neuroendocrine carcinomas. PubMed, Cochrane Library, and Ichushi-Web databases were searched for information sources. Independent systematic reviewers conducted two rounds of screening and selected relevant records for each CQ. Finally, the working group members synthesized the strength of evidence and recommendations. RESULTS After two rounds of screening, 5/0/3/0/2/0 records were extracted for CQ1 of esophageal/gastric/pancreatic/biliary tract/colorectal/ and neuroendocrine carcinoma, respectively. Additionally, a total of 2/6/1 records were extracted for CQ2 of esophageal/pancreatic/colorectal cancer, respectively. The strength of evidence and recommendations were evaluated for CQ1 of colorectal cancer; however, we could not synthesize recommendations for other CQs owing to the lack of records. CONCLUSION The use of G-CSF for primary prophylaxis in chemotherapy for colorectal cancer is inappropriate.
Collapse
Affiliation(s)
- Mamoru Ito
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuta Okumura
- Department of Gastrointestinal and Medical Oncology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Kenta Nio
- Department of Medical Oncology, Hamanomachi Hospital, Fukuoka, Japan
| | - Eishi Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yukinori Ozaki
- Department of Breast Medical Oncology, The Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroshi Nishio
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Eiki Ichihara
- Center for Clinical Oncology, Okayama University Hospital, Okayama, Japan
| | - Yuji Miura
- Department of Medical Oncology, Toranomon Hospital, Tokyo, Japan
| | - Makoto Endo
- Department of Orthopaedic Surgery, Kyushu University, Fukuoka, Japan
| | - Shingo Yano
- Division of Clinical Oncology/Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Dai Maruyama
- Department of Hematology Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tetsuhiro Yoshinami
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Nobuyuki Susumu
- Department of Obstetrics and Gynecology, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | | | - Takashi Motohashi
- Department of Obstetrics and Gynecology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Nobuaki Ochi
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Toshio Kubo
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Keita Uchino
- Department of Medical Oncology, NTT Medical Center Tokyo, Tokyo, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yutaro Kamiyama
- Department of Clinical Oncology/Hematology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shinji Nakao
- Department of Hematology, Faculty of Medicine, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Shinobu Tamura
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Hitomi Nishimoto
- Department of Nursing, Okayama University Hospital, Okayama, Japan
| | - Yasuhisa Kato
- Department of Drug Information, Faculty of Pharmaceutical Sciences, Shonan University of Medical Sciences, Kanagawa, Japan
| | - Atsushi Sato
- Department of Medical Oncology, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Toshimi Takano
- Department of Breast Medical Oncology, The Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kenji Tsuchihashi
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
404
|
Yamaguchi J, Kokuryo T, Yokoyama Y, Oishi S, Sunagawa M, Mizuno T, Onoe S, Watanabe N, Ogura A, Ebata T. Trefoil factor 1 suppresses stemness and enhances chemosensitivity of pancreatic cancer. Cancer Med 2024; 13:e7395. [PMID: 38872370 PMCID: PMC11176577 DOI: 10.1002/cam4.7395] [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: 11/12/2023] [Revised: 05/30/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND AND AIMS Pancreatic cancer is one of the most lethal malignancies, partly due to resistance to conventional chemotherapy. The chemoresistance of malignant tumors is associated with epithelial-mesenchymal transition (EMT) and the stemness of cancer cells. The aim of this study is to investigate the availability and functional mechanisms of trefoil factor family 1 (TFF1), a tumor-suppressive protein in pancreatic carcinogenesis, to treat pancreatic cancer. METHODS To investigate the role of endogenous TFF1 in human and mice, specimens of human pancreatic cancer and genetically engineered mouse model of pancreatic cancer (KPC/TFF1KO; Pdx1-Cre/LSL-KRASG12D/LSL-p53R172H/TFF1-/-) were analyzed by immunohistochemistry (IHC). To explore the efficacy of extracellular administration of TFF1, recombinant and chemically synthesized TFF1 were administered to pancreatic cancer cell lines, a xenograft mouse model and a transgenic mouse model. RESULTS The deficiency of TFF1 was associated with increased EMT of cancer cells in mouse models of pancreatic cancer, KPC. The expression of TFF1 in cancer cells was associated with better survival rate of the patients who underwent chemotherapy, and loss of TFF1 deteriorated the benefit of gemcitabine in KPC mice. Extracellular administration of TFF1 inhibited gemcitabine-induced EMT, Wnt pathway activation and cancer stemness, eventually increased apoptosis of pancreatic cancer cells in vitro. In vivo, combined treatment of gemcitabine and subcutaneous administration of TFF1 arrested tumor growth in xenograft mouse model and resulted in the better survival of KPC mice by inhibiting EMT and cancer stemness. CONCLUSION These results indicate that TFF1 can contribute to establishing a novel strategy to treat pancreatic cancer patients by enhancing chemosensitivity.
Collapse
Affiliation(s)
- Junpei Yamaguchi
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshio Kokuryo
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Yokoyama
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shunsuke Oishi
- Institute of Transformative Bio-Molecules, Nagoya University, Nagoya, Japan
| | - Masaki Sunagawa
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Mizuno
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shunsuke Onoe
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuyuki Watanabe
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Ogura
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoki Ebata
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
405
|
Kosinski J, Sechi A, Hain J, Villwock S, Ha SA, Hauschulz M, Rose M, Steib F, Ortiz‐Brüchle N, Heij L, Maas SL, van der Vorst EPC, Knoesel T, Altendorf‐Hofmann A, Simon R, Sauter G, Bednarsch J, Jonigk D, Dahl E. ITIH5 as a multifaceted player in pancreatic cancer suppression, impairing tyrosine kinase signaling, cell adhesion and migration. Mol Oncol 2024; 18:1486-1509. [PMID: 38375974 PMCID: PMC11161730 DOI: 10.1002/1878-0261.13609] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/19/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024] Open
Abstract
Inter-alpha-trypsin inhibitor heavy chain 5 (ITIH5) has been identified as a metastasis suppressor gene in pancreatic cancer. Here, we analyzed ITIH5 promoter methylation and protein expression in The Cancer Genome Atlas (TCGA) dataset and three tissue microarray cohorts (n = 618), respectively. Cellular effects, including cell migration, focal adhesion formation and protein tyrosine kinase activity, induced by forced ITIH5 expression in pancreatic cancer cell lines were studied in stable transfectants. ITIH5 promoter hypermethylation was associated with unfavorable prognosis, while immunohistochemistry demonstrated loss of ITIH5 in the metastatic setting and worsened overall survival. Gain-of-function models showed a significant reduction in migration capacity, but no alteration in proliferation. Focal adhesions in cells re-expressing ITIH5 exhibited a smaller and more rounded phenotype, typical for slow-moving cells. An impressive increase of acetylated alpha-tubulin was observed in ITIH5-positive cells, indicating more stable microtubules. In addition, we found significantly decreased activities of kinases related to focal adhesion. Our results indicate that loss of ITIH5 in pancreatic cancer profoundly affects its molecular profile: ITIH5 potentially interferes with a variety of oncogenic signaling pathways, including the PI3K/AKT pathway. This may lead to altered cell migration and focal adhesion formation. These cellular alterations may contribute to the metastasis-inhibiting properties of ITIH5 in pancreatic cancer.
Collapse
Affiliation(s)
- Jennifer Kosinski
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Antonio Sechi
- Department of Cell and Tumor BiologyRWTH Aachen UniversityGermany
| | - Johanna Hain
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Sophia Villwock
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Stefanie Anh Ha
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Maximilian Hauschulz
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Michael Rose
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Florian Steib
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Nadina Ortiz‐Brüchle
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Lara Heij
- Institute of PathologyUniversity Hospital EssenGermany
- Department of Surgery and Transplantation, Medical FacultyRWTH Aachen UniversityGermany
- Department of PathologyErasmus Medical Center RotterdamThe Netherlands
- NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht UniversityThe Netherlands
| | - Sanne L. Maas
- Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR)Medical Faculty of RWTH Aachen UniversityGermany
| | - Emiel P. C. van der Vorst
- Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR)Medical Faculty of RWTH Aachen UniversityGermany
- Institute for Cardiovascular Prevention (IPEK)Ludwig‐Maximilians‐University MunichGermany
| | - Thomas Knoesel
- Institute of PathologyLudwig‐Maximilians‐University MunichGermany
| | | | - Ronald Simon
- Institute of PathologyUniversity Medical Center Hamburg‐EppendorfGermany
| | - Guido Sauter
- Institute of PathologyUniversity Medical Center Hamburg‐EppendorfGermany
| | - Jan Bednarsch
- Department of Surgery and Transplantation, Medical FacultyRWTH Aachen UniversityGermany
| | - Danny Jonigk
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- RWTH centralized Biomaterial Bank (RWTH cBMB)Medical Faculty of the RWTH Aachen UniversityGermany
- German Center for Lung Research (DZL), BREATHHanoverGermany
| | - Edgar Dahl
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- RWTH centralized Biomaterial Bank (RWTH cBMB)Medical Faculty of the RWTH Aachen UniversityGermany
| |
Collapse
|
406
|
Sahin TK, Ozbay Y, Altunbulak AY, Altunbulak HI, Onur MR, Ceylan F, Guven DC, Yalcin S, Dizdar O. Albumin-myosteatosis gauge as a prognostic factor in patients with advanced pancreatic cancer undergoing first-line chemotherapy. Int J Clin Oncol 2024; 29:822-831. [PMID: 38565751 DOI: 10.1007/s10147-024-02512-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Sarcopenia and myosteatosis have been associated with a poor prognosis for several cancers. The albumin-myosteatosis gauge (AMG) is a novel integrated measure proposed to assess myosteatosis along with serum albumin level as a surrogate of systemic inflammation and malnutrition. The aim of this study was to investigate the prognostic value of AMG in patients with advanced pancreatic ductal adenocarcinoma (PDAC). METHODS Patients with advanced PDAC treated with chemotherapy between 2013 and 2022 were evaluated. Skeletal muscle radiodensity (SMD) and skeletal muscle index (SMI) were calculated using computed tomography at the level of the L3 vertebra. The AMG was defined as albumin x SMD and expressed as an arbitrary unit (AU). Patients were first categorized by sex-specific quartiles and then dichotomized at the sex-specific median value of the AMG. RESULTS A total of 196 patients were included. The median age (interquartile range) was 62 (54-67), and 128 (65.3%) were male. With regard to AMG, 142.86 and 114.15 AU were identified as cutoff values for males and females, respectively. In multivariable analyses, lower AMG values (G1-G2 vs. G3-G4) (HR: 1.61, 95% CI 1.17-2.21, p = 0.003), higher ECOG performance score (> 0 vs. 0) (HR: 1.51, 95% CI 1.10-2.06, p = 0.009) and metastatic disease (vs. locally advanced) (HR: 1.88, 95% CI 1.27-2.79, p = 0.001) were associated with OS. CONCLUSION The study findings suggest the prognostic value of AMG in patients with advanced PDAC undergoing first-line chemotherapy. Further studies are warranted to validate these findings and assess potential predictive role of AMG in guiding treatment selection.
Collapse
Affiliation(s)
- Taha Koray Sahin
- Department of Medical Oncology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100, Ankara, Turkey.
| | - Yakup Ozbay
- Department of Radiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | | | | | - Mehmet Ruhi Onur
- Department of Radiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Furkan Ceylan
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Deniz Can Guven
- Department of Medical Oncology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Suayib Yalcin
- Department of Medical Oncology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Omer Dizdar
- Department of Medical Oncology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| |
Collapse
|
407
|
Giulia O, Alessandro B, Angelo C, Paolo M, Rosa C, Marina M, Umberto P, Catia C, Giulia V, Massimo F, Michele R. Isolated lung metastases from pancreatic ductal adenocarcinoma (PDAC): Diagnostic and therapeutic challenges of a different disease. Semin Oncol 2024; 51:69-76. [PMID: 38879350 DOI: 10.1053/j.seminoncol.2024.04.001] [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: 09/28/2023] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 08/04/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, mostly due to the high rate of distant dissemination. However, growing evidence shows that isolated lung recurrence or metastases (ILM) from PDAC are not only less common, but also correlated with a better prognosis. Lung-only recurrence after surgery occurs later in time and is associated with more favorable prognostic characteristics of the primary tumor. Moreover, recent findings suggest that this specific site of metastases is characterized by an immunologically "hot" microenvironment and a more favorable molecular profile that could possibly justify its clinical behavior. Thus, ILM from PDAC emerge as a distinct entity, that might also benefit from a different therapeutic approach, possibly with the integration of surgery and de-intensified chemotherapy regimens, especially in selected patients. In this review we delve into the current scientific evidence on the clinical and biological characteristics of isolated LM from PDAC, also focusing on concerns with their diagnostic process and the therapeutic options for the management of this subset of patients.
Collapse
Affiliation(s)
- Orsi Giulia
- Department of Medical Oncology, Vita-Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Bandiera Alessandro
- Department of Thoracic Surgery, Vita- Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carretta Angelo
- Department of Thoracic Surgery, Vita- Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Migliori Paolo
- Medical Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Chavez Rosa
- Department of Medical Oncology, Vita-Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Macchini Marina
- Department of Medical Oncology, Vita-Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Peretti Umberto
- Department of Medical Oncology, Vita-Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carconi Catia
- Department of Medical Oncology, Vita-Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Veronesi Giulia
- Department of Thoracic Surgery, Vita- Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Falconi Massimo
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Centre, Vita -Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Reni Michele
- Department of Medical Oncology, Vita-Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
408
|
Koti S, Demyan L, Deutsch G, Weiss M. Surgery for Oligometastatic Pancreatic Cancer: Defining Biologic Resectability. Ann Surg Oncol 2024; 31:4031-4041. [PMID: 38502293 PMCID: PMC11076395 DOI: 10.1245/s10434-024-15129-8] [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: 12/14/2023] [Accepted: 02/17/2024] [Indexed: 03/21/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is most often metastatic at diagnosis. As systemic therapy continues to improve alongside advanced surgical techniques, the focus has shifted toward defining biologic, rather than technical, resectability. Several centers have reported metastasectomy for oligometastatic PDAC, yet the indications and potential benefits remain unclear. In this review, we attempt to define oligometastatic disease in PDAC and to explore the rationale for metastasectomy. We evaluate the existing evidence for metastasectomy in liver, peritoneum, and lung individually, assessing the safety and oncologic outcomes for each. Furthermore, we explore contemporary biomarkers of biological resectability in oligometastatic PDAC, including radiographic findings, biochemical markers (such as CA 19-9 and CEA), inflammatory markers (including neutrophil-to-lymphocyte ratio, C-reactive protein, and scoring indices), and liquid biopsy techniques. With careful consideration of existing data, we explore the concept of biologic resectability in guiding patient selection for metastasectomy in PDAC.
Collapse
Affiliation(s)
- Shruti Koti
- Department of General Surgery, Northwell Health, Queens, NY, USA.
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY, USA.
| | - Lyudmyla Demyan
- Department of General Surgery, Northwell Health, Queens, NY, USA
| | - Gary Deutsch
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Matthew Weiss
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| |
Collapse
|
409
|
Singh I, Chou JF, Capanu M, Park J, Yu KH, Varghese AM, Park W, Zervoudakis A, Keane F, Rolston VS, Gerdes H, Wei AC, Shah P, Covey A, Schattner M, O'Reilly EM. Morbidity and mortality in patients with stage IV pancreatic adenocarcinoma and acute cholangitis: Outcomes and risk prognostication. Pancreatology 2024; 24:608-615. [PMID: 38749803 PMCID: PMC11164623 DOI: 10.1016/j.pan.2024.05.515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Acute cholangitis (AC) is a common complication of pancreatic ductal adenocarcinoma (PDAC). Herein, we evaluated outcomes after the first AC episode and predictors of mortality and AC recurrence in patients with stage IV PDAC. METHODS We conducted a single-center, retrospective observational study using institutional databases. Clinical data and outcomes for patients with stage IV PDAC and at least one documented episode of AC, were assessed. Overall survival (OS) was estimated using the Kaplan-Meier method, and Cox regression model was employed to identify predictors of AC recurrence and mortality. RESULTS One hundred and twenty-four patients with stage IV PDAC and AC identified between January 01, 2014 and October 31, 2020 were included. Median OS after first episode of AC was 4.1 months (95 % CI, 4.0-5.5), and 30-day, 6, and 12-month survival was 86.2 % (95 % CI, 80.3-92.5), 37 % (95 % CI, 29.3-46.6 %) and 18.9 % (95 % CI, 13.1-27.3 %), respectively. Primary tumor in pancreatic body/tail (HR 2.29, 95 % CI: 1.26 to 4.18, p = 0.011), concomitant metastases to liver and other sites (HR 1.96, 95 % CI: 1.16 to 3.31, p = 0.003) and grade 3 AC (HR 2.26, 95 % CI: 1.45 to 3.52, p < 0.001), predicted worse outcomes. Intensive care unit admission, sepsis, systemic therapy, treatment regimen, and time to intervention did not predict survival or risk of recurrence of AC. CONCLUSIONS AC confers significant morbidity and mortality in advanced PDAC. Worse outcomes are associated with higher grade AC, primary tumor location in pancreatic body/tail, and metastases to liver and other sites.
Collapse
Affiliation(s)
- Isha Singh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Medicine, West Virginia University School of Medicine, Morgantown, WV, 26505, USA
| | - Joanne F Chou
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Marinela Capanu
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jennifer Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Kenneth H Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Anna M Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Alice Zervoudakis
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Fergus Keane
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Vineet Syan Rolston
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Hans Gerdes
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Alice C Wei
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Pari Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Anne Covey
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Mark Schattner
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
410
|
Wattenberg MM, Colby S, Garrido-Laguna I, Xue Y, Chang R, Delman D, Lee J, Affolter K, Mulvihill SJ, Beg MS, Wang-Gillam A, Wade JL, Guthrie KA, Chiorean EG, Ahmad SA, Lowy AM, Philip PA, Sohal DPS, Beatty GL. Intratumoral Cell Neighborhoods Coordinate Outcomes in Pancreatic Ductal Adenocarcinoma. Gastroenterology 2024; 166:1114-1129. [PMID: 38244727 PMCID: PMC11102852 DOI: 10.1053/j.gastro.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDA) is a highly lethal disease characterized by a spatially heterogeneous tumor microenvironment. Within the PDA microenvironment, cells organize into communities where cell fate is influenced by neighboring cells of diverse ontogeny and function. However, it remains unclear how cell neighborhoods in the tumor microenvironment evolve with treatment and impact clinical outcomes. METHODS Here, using automated chromogenic multiplex immunohistochemistry and unsupervised computational image analysis of human PDA tumors, we investigated cell neighborhoods in surgically resected tumors from patients with chemotherapy-naïve PDA (n = 59) and neoadjuvant chemotherapy-treated PDA (n = 57). Single cells were defined by lineage markers (CD3, CD8, Foxp3, CD68, CK19), proliferation (Ki67), and neighboring cells. RESULTS Distinct intratumoral immune and tumor cell subsets were defined by neighboring cells. Higher content of stromal-associated macrophages was seen in chemotherapy-naïve tumors from long-term survivors (overall survival >3 years) compared with short-term survivors (overall survival <1 year), whereas immune-excluded tumor cells were higher in short-term survivors. Chemotherapy-treated vs -naïve tumors showed lower content of tumor-associated T cells and macrophages but similar densities of stromal-associated immune cells. However, proliferating tumor cell subsets with immune-rich neighborhoods were higher in chemotherapy-treated tumors. In a blinded analysis of tumors from patients treated with neoadjuvant chemotherapy, a composite index comprising lower quantities of immune-excluded tumor cells and higher spatially distinct immune cell subsets was associated with prolonged survival. CONCLUSIONS Together, these data provide new insights into discrete cell communities in PDA and show their clinical relevance.
Collapse
Affiliation(s)
- Max M Wattenberg
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sarah Colby
- SWOG Statistics and Data Management Center, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Ignacio Garrido-Laguna
- Division of Oncology, Department of Internal Medicine, University of Utah School of Medicine, University of Utah, Salt Lake City, Utah
| | - Yuqing Xue
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Renee Chang
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Devora Delman
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jesse Lee
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kajsa Affolter
- Department of Pathology, University of Utah School of Medicine, University of Utah, Salt Lake City, Utah
| | - Sean J Mulvihill
- Department of Surgery, University of Utah School of Medicine, University of Utah, Salt Lake City, Utah
| | | | | | | | - Katherine A Guthrie
- SWOG Statistics and Data Management Center, Fred Hutchinson Cancer Center, Seattle, Washington
| | - E Gabriela Chiorean
- University of Washington, Fred Hutchinson Cancer Center, Seattle, Washington
| | | | - Andrew M Lowy
- Division of Surgical Oncology, Department of Surgery, UC San Diego, La Jolla, California
| | - Philip Agop Philip
- Henry Ford Health, Wayne State University, Oncology and Pharmacology, Detroit, Michigan
| | | | - Gregory L Beatty
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
411
|
Imamura T, Ohgi K, Mori K, Ashida R, Yamada M, Otsuka S, Uesaka K, Sugiura T. Surrogacy of Recurrence-free Survival for Overall Survival as an Endpoint of Clinical Trials of Perioperative Adjuvant Therapy in Hepatobiliary-pancreatic Cancers: A Retrospective Study and Meta-analysis. Ann Surg 2024; 279:1025-1035. [PMID: 37638472 DOI: 10.1097/sla.0000000000006084] [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: 08/29/2023]
Abstract
OBJECTIVE To assess the correlation between recurrence-free survival (RFS) and overall survival (OS) in the hepatobiliary-pancreatic (HBP) surgical setting to validate RFS as a surrogate endpoint. BACKGROUND Reliable surrogate endpoints for OS are still limited in the field of HBP surgery. METHODS We analyzed patients who underwent curative resection for HBP disease [986 patients with pancreatic ductal adenocarcinoma (PDAC), 1168 with biliary tract cancer (BTC), 1043 with hepatocellular carcinoma, and 1071 with colorectal liver metastasis] from September 2002 to June 2022. We also conducted meta-analyses of randomized controlled trials of neoadjuvant or adjuvant therapy to validate the surrogacy in PDAC and BTC. RESULTS Correlation coefficients between RFS and OS were low for hepatocellular carcinoma ( p = 0.67) and colorectal liver metastasis ( p = 0.53) but strong for PDAC ( p = 0.80) and BTC ( p = 0.75). In a landmark analysis, the concordance rates between survival or death at 5 years postoperatively and the presence or absence of recurrence at each time point (1, 2, 3, and 4 years) were 50%, 70%, 74%, and 77% for PDAC and 54%, 67%, 73%, and 78% for BTC, respectively, both increasing and reaching a plateau at 3 years. In a meta-analysis, the correlation coefficients for the RFS hazard ratio and OS hazard ratio in PDAC and BTC were p = 0.88 ( P < 0.001) and p = 0.87 ( P < 0.001), respectively. CONCLUSIONS Three-year RFS can be a reliable surrogate endpoint for OS in clinical trials of neoadjuvant or adjuvant therapy for PDAC and BTC.
Collapse
Affiliation(s)
- Taisuke Imamura
- Division of HepatoBiliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Katsuhisa Ohgi
- Division of HepatoBiliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Keita Mori
- Clinical Research Center, Shizuoka Cancer Center, Shizuoka, Japan
| | - Ryo Ashida
- Division of HepatoBiliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Mihoko Yamada
- Division of HepatoBiliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Shimpei Otsuka
- Division of HepatoBiliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Katsuhiko Uesaka
- Division of HepatoBiliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Teiichi Sugiura
- Division of HepatoBiliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
412
|
Haberkorn BCM, Hoogendijk L, Loosveld OS, Thijs AMJ, Verstijnen J. Real Life Data and Outcome of FOLFIRINOX Use in Metastatic Pancreatic Cancer Patients in General Hospitals in the Netherlands. J Gastrointest Cancer 2024; 55:838-844. [PMID: 38319559 DOI: 10.1007/s12029-023-01006-2] [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: 12/21/2023] [Indexed: 02/07/2024]
Abstract
INTRODUCTION Pancreatic cancer is one of the five most common causes of cancer mortality in developed countries. In patients with metastatic disease, the most frequent treatment used is FOLFIRINOX, which is associated with moderate toxicity which could influence quality of life. The efficacy of FOLFIRINOX in a general population in the Netherlands has not been subject of research before, and therefore, this research has been set up in order to investigate what the real-life benefits of FOLFIRINOX are in a population with metastatic pancreatic cancer (mPC) treated in three general hospitals in the Netherlands. METHODS The data used in this study was collected by patient records leading to a noninterventional retrospective cohort study. Eighty-six patients, over 18 years of age, diagnosed with mPC between the years 2015 and 2022 and treated with FOLFIRINOX at Maasstad Hospital in Rotterdam, Amphia Hospital in Breda, or Catharina Hospital in Eindhoven, were included in the study. Kaplan-Meier models were used in order to represent survival outcomes. RESULTS The results showed a median overall survival of 228 days (IQR 118-355). Only 14.0% (n = 12) completed the first-line treatment, and 51.2% (n = 44) of patients stopped treatment before or during cycle 6. Toxicity is highest, grade 3, after the first cycle but remains high for grade 1 and 2 during all treatment cycles. CONCLUSION Survival rates for patient with metastatic pancreatic cancer treated with FOLFIRINOX were worse in our study population than in comparative studies.
Collapse
Affiliation(s)
- B C M Haberkorn
- Department of Medical Oncology, Maasstad Ziekenhuis Rotterdam, Maasstadweg 21, 3079 DZ, Rotterdam, the Netherlands.
| | | | - O S Loosveld
- Department of Medical Oncology, Amphia Ziekenhuis Breda, Breda, Netherlands
| | - A M J Thijs
- Department of Medical Oncology, Catharina Ziekenhuis Eindhoven, Eindhoven, Netherlands
| | - J Verstijnen
- Department of Medical Oncology, Maasstad Ziekenhuis Rotterdam, Maasstadweg 21, 3079 DZ, Rotterdam, the Netherlands
| |
Collapse
|
413
|
Giordano G, Milella M, Landriscina M, Bergamo F, Tirino G, Santaniello A, Zaniboni A, Vasile E, De Vita F, Re GL, Vaccaro V, Giommoni E, Natale D, Conca R, Santini D, Maiorino L, Sanna G, Ricci V, Iop A, Montesarchio V, Procaccio L, Noventa S, Bianco R, Febbraro A, Lonardi S, Tortora G, Sperduti I, Melisi D. Prognostic analysis and outcomes of metastatic pancreatic cancer patients receiving nab-paclitaxel plus gemcitabine as second or later-line treatment. Cancer Med 2024; 13:e7345. [PMID: 38924262 PMCID: PMC11199338 DOI: 10.1002/cam4.7345] [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: 01/24/2024] [Revised: 05/11/2024] [Accepted: 05/18/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Pancreatic cancer (PC) first-line therapy often consists of polychemotherapy regimens, but choosing a second-line therapy after disease progression, especially following first-line FOLFIRINOX, remains a clinical challenge. This study presents results from a large, multicenter, retrospective analysis of Italian patients with metastatic PC (mPC) treated with Nab-paclitaxel/Gemcitabine (AG) as second or later line of treatment. Main objective of the study is to identify prognostic factors that could inform treatment decisions. METHODS The study included 160 mPC patients treated with AG in 17 Italian institutions. AG was administered according to labelling dose, until disease progression, unacceptable toxicity or patient refusal. Variations in schedules, dose modifications, supportive measures, and response evaluation were determined by individual clinicians' practice. RESULTS AG was well-tolerated and exhibited promising clinical activity. The overall response rate (ORR) and the disease control rate (DCR) were 22.5% and 45.6%, respectively. Median progression-free survival (PFS) and overall survival (OS) were 3.9 and 6.8 months, respectively. Among the patients who received AG as a second-line therapy (n = 111, 66.9%), median PFS and OS were 4.2 and 7.4 months, respectively. Notably, in the 76 patients (68%) receiving AG after first-line FOLFIRINOX, an ORR of 19.7% and a DCR of 46.0% were observed, resulting in a median PFS of 3.5 and median OS of 5.7 months. The study identified specific clinical or laboratory parameters (LDH, NLR, fasting serum glucose, liver metastases, ECOG PS, and first-line PFS) as independent prognostic factors at multivariate level. These factors were used to create a prognostic nomogram that divided patients into three risk classes, helping to predict second-line OS and PFS. CONCLUSIONS This study represents the largest real-world population of mPC patients treated with AG as a second or later line of therapy. It supports the feasibility of this regimen following first-line FOLFIRINOX, particularly in patients with specific clinical and laboratory characteristics who derived prolonged benefit from first-line therapy.
Collapse
Affiliation(s)
- Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical SciencesUniversity of FoggiaFoggiaItaly
| | - Michele Milella
- Section of Oncology, Department of MedicineUniversity of Verona School of Medicine and Verona University Hospital TrustVeronaItaly
| | - Matteo Landriscina
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical SciencesUniversity of FoggiaFoggiaItaly
| | | | - Giuseppe Tirino
- Unit of Medical Oncology, Sacro Cuore di Gesu'—Fatebenefratelli HospitalBeneventoItaly
| | - Antonio Santaniello
- Department of Clinical Medicine and SurgeryUniversity of Naples "Federico II"NaplesItaly
| | | | - Enrico Vasile
- Unit of Medical Oncology 2Azienda Ospedaliero‐Universitaria PisanaPisaItaly
| | - Ferdinando De Vita
- Division of Medical Oncology, Department of Precision MedicineUniversity of Campania "L. Vanvitelli"NaplesItaly
| | - Giovanni Lo Re
- Medical Oncology and Immune‐Related TumorsCentro di Riferimento Oncologico di Aviano (CRO), IRCCSAvianoItaly
| | - Vanja Vaccaro
- Medical Oncology 1IRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Elisa Giommoni
- Medical Oncology UnitCareggi University HospitalFlorenceItaly
| | | | - Raffaele Conca
- Division of Medical Oncology, Department of Onco‐HematologyIRCCS‐CROB, Referral Cancer Center of BasilicataRionero in VultureItaly
| | - Daniele Santini
- Medical Oncology AUniversity of Rome, Policlinico Umberto I, "La SapienzaRomeItaly
| | | | - Gianni Sanna
- Medical OncologyIstituto Ospedaliero dell'Università di SassariSassariItaly
| | - Vincenzo Ricci
- Medical Oncology UnitAzienda Ospedaliera di Rilievo Nazionale ‘San Pio’BeneventoItaly
| | - Aldo Iop
- Department of OncologyAzienda Sanitaria Universitaria Giuliano Isontina (ASUGI)TriesteItaly
| | | | | | - Silvia Noventa
- Medical Oncology UnitFondazione PoliambulanzaBresciaItaly
| | - Roberto Bianco
- Department of Clinical Medicine and SurgeryUniversity of Naples "Federico II"NaplesItaly
| | - Antonio Febbraro
- Unit of Medical Oncology, Sacro Cuore di Gesu'—Fatebenefratelli HospitalBeneventoItaly
| | - Sara Lonardi
- Department of OncologyVeneto Institute of Oncology IRCCSPadovaItaly
| | - Giampaolo Tortora
- Oncologia MedicaFondazione Policlinico Universitario Gemelli IRCCSRomeItaly
- Oncologia MedicaUniversità Cattolica del Sacro CuoreRomeItaly
| | - Isabella Sperduti
- Biostatistical UnitIRCCS Regina Elena National Cancer Institute, Istituti Fisioterapici OspitalieriRomeItaly
| | - Davide Melisi
- Section of Oncology, Department of MedicineUniversity of Verona School of Medicine and Verona University Hospital TrustVeronaItaly
- Investigational Cancer Therapeutics Clinical UnitAzienda Ospedaliera Universitaria IntegrataVeronaItaly
- Digestive Molecular Clinical Oncology Research UnitUniversity of VeronaVeronaItaly
| |
Collapse
|
414
|
Ohyama H, Hirotsu Y, Amemiya K, Amano H, Hirose S, Oyama T, Iimuro Y, Kojima Y, Mikata R, Mochizuki H, Kato N, Omata M. Liquid biopsy of wash samples obtained via endoscopic ultrasound-guided fine-needle biopsy: Comparison with liquid biopsy of plasma in pancreatic cancer. Diagn Cytopathol 2024; 52:325-331. [PMID: 38516904 DOI: 10.1002/dc.25306] [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: 02/17/2024] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
OBJECTIVES Pancreatic cancer (PC) has a poor prognosis and limited treatment options. Liquid biopsy, which analyzes circulating tumor DNA (ctDNA) in blood, holds promise for precision medicine; however, low ctDNA detection rates pose challenges. This study aimed to investigate the utility of wash samples obtained via endoscopic ultrasound-guided fine-needle biopsy (EUS-FNB) as a liquid biopsy for PC. METHODS A total of 166 samples (42 formalin-fixed paraffin-embedded [FFPE] tissues, 80 wash samples, and 44 plasma samples) were collected from 48 patients with PC for genomic analysis. DNA was extracted and quantified, and 60 significantly mutated genes were sequenced. The genomic profiles of FFPE tissues, wash samples, and plasma samples were compared. Finally, the ability to detect druggable mutations in 80 wash samples and 44 plasma samples was investigated. RESULTS The amount of DNA was significantly lower in plasma samples than in wash samples. Genomic analysis revealed a higher detection rate of oncogenic mutations in FFPE tissues (98%) and wash samples (96%) than in plasma samples (18%) and a comparable detection rate in FFPE tissues and wash samples. Tumor-derived oncogenic mutations were detected more frequently in wash samples than in plasma samples. Furthermore, the oncogenic mutations detection rate remained high in wash samples at all PC stages but low in plasma samples even at advanced PC stages. Using wash samples was more sensitive than plasma samples for identifying oncogenic and druggable mutations. CONCLUSIONS The wash sample obtained via EUS-FNB is an ideal specimen for use as a liquid biopsy for PC.
Collapse
Affiliation(s)
- Hiroshi Ohyama
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Genome Analysis Center, Yamanashi Central Hospital, Yamanashi, Japan
- Department of Gastroenterology, Yamanashi Central Hospital, Yamanashi, Japan
| | - Yosuke Hirotsu
- Genome Analysis Center, Yamanashi Central Hospital, Yamanashi, Japan
| | - Kenji Amemiya
- Genome Analysis Center, Yamanashi Central Hospital, Yamanashi, Japan
| | - Hiroyuki Amano
- Department of Gastroenterology, Yamanashi Central Hospital, Yamanashi, Japan
| | - Sumio Hirose
- Department of Gastroenterology, Yamanashi Central Hospital, Yamanashi, Japan
| | - Toshio Oyama
- Department of Pathology, Yamanashi Central Hospital, Yamanashi, Japan
| | - Yuji Iimuro
- Department of Surgery, Yamanashi Central Hospital, Yamanashi, Japan
| | - Yuichiro Kojima
- Department of Gastroenterology, Yamanashi Central Hospital, Yamanashi, Japan
| | - Rintaro Mikata
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hitoshi Mochizuki
- Genome Analysis Center, Yamanashi Central Hospital, Yamanashi, Japan
- Department of Gastroenterology, Yamanashi Central Hospital, Yamanashi, Japan
| | - Naoya Kato
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masao Omata
- Genome Analysis Center, Yamanashi Central Hospital, Yamanashi, Japan
- Department of Gastroenterology, Yamanashi Central Hospital, Yamanashi, Japan
- University of Tokyo, Tokyo, Japan
| |
Collapse
|
415
|
Fan M, Ma Y, Deng G, Si H, Jia R, Wang Z, Dai G. A real-world analysis of second-line treatment option, gemcitabine plus anlotinib and anti-PD1, in advanced pancreatic cancer. Pancreatology 2024; 24:579-583. [PMID: 38553260 DOI: 10.1016/j.pan.2024.03.017] [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: 02/19/2023] [Revised: 03/04/2024] [Accepted: 03/23/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND In the second-line treatment of advanced pancreatic cancer (APC), there is only one approved regimen based on the phase III NAPOLI-1 trial. However, for patients progressing after Nab-paclitaxel and Gemcitabine (Nab-P/Gem) or Nab-P combinations, second-line treatment were very limited. METHODS This is a retrospective single-center analysis of patients. Our aim was to determine the effectiveness and tolerability of a novel regimen, gemcitabine plus Anlotinib and anti-PD1, in APC patients and to compare it with oxaliplatin, irinotecan, leucovorin, and fluorouracil (FOLFIRINOX) in the second-line setting who have failed on the first-line Nab-P combinations. RESULTS In total, twenty-three patients received Gemcitabine plus Anlotinib and anti-PD1 in the second-line, 28 patients were treated with FOLFORINOX. There was no significant difference in overall survival (OS) or progression free survival (PFS) for either of the two sequences (p > 0.05). Patients who received Gemcitabine plus Anlotinib and anti-PD1 had a median PFS of 4.0 months (95% CI: 1.1-6.9) versus 3.5 months (95% CI 1.8-5.2) in FOLFORINOX group (p = 0.953). The median OS of Gemcitabine plus Anlotinib and anti-PD1 was 9.0 months (95% CI: 4.0-13.7) and 8.0 months (95% CI: 5.5-10.5) in FOLFORINOX group (p = 0.373). Grade ≥3 treatment-emergent adverse events (AEs) occurred for 13% of patients with Gemcitabine plus Anlotinib and anti-PD1 and 40% for FOLFORINOX. CONCLUSION Our data confirms the effectiveness of Gemcitabine plus Anlotinib and anti-PD1 as a well-tolerated regimen in the second-line treatment of APC and extends available data on its use as a second-line treatment option when compared with FOLFIRINOX.
Collapse
Affiliation(s)
- Mengjiao Fan
- Medical School of Chinese People's Liberation Army, Beijing, China; Medical Oncology Department, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China; Medical Oncology Department, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Yue Ma
- Medical School of Chinese People's Liberation Army, Beijing, China; Medical Oncology Department, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China; Medical Oncology Department, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Guochao Deng
- Medical Oncology Department, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Haiyan Si
- Medical Oncology Department, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Ru Jia
- Medical Oncology Department, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Zhikuan Wang
- Medical Oncology Department, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Guanghai Dai
- Medical Oncology Department, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China; Medical Oncology Department, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.
| |
Collapse
|
416
|
Christensen TD, Maag E, Theile S, Madsen K, Lindgaard SC, Hasselby JP, Nielsen DL, Johansen JS, Chen IM. Circulating immune-related proteins associated with immune checkpoint inhibitor efficacy in patients with pancreatic ductal adenocarcinoma. ESMO Open 2024; 9:103489. [PMID: 38838501 PMCID: PMC11190466 DOI: 10.1016/j.esmoop.2024.103489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/02/2024] [Accepted: 05/02/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Most patients with pancreatic ductal adenocarcinoma (PDAC) do not benefit from immune checkpoint inhibitor treatment. However, the phase II study CheckPAC (NCT02866383) showed a clinical benefit (CB) rate of 37% and a response rate of 14% in patients with metastatic PDAC receiving stereotactic radiation therapy and nivolumab with or without ipilimumab. Translational studies were initiated to characterize the patients who would benefit from this treatment. Here, we evaluated the association between treatment outcome and 92 circulating immuno-oncology-related proteins in patients from the CheckPAC trial. MATERIALS AND METHODS The study included 78 patients with chemoresistant metastatic PDAC treated with nivolumab ± ipilimumab combined with radiotherapy. Proteins were measured in serum samples collected at baseline and on treatment with the use of the Olink Target 96 Immuno-Oncology panel. A cohort of 234 patients with metastatic PDAC treated with first-line chemotherapy were also included. RESULTS High levels of Fas ligand (FASLG) and galectin 1 (Gal-1) and low levels of C-C motif chemokine 4 were associated with CB. High FASLG and Gal-1 were associated with longer progression-free survival in univariable analysis. In the multivariable Cox regression analysis, the association was significant for Gal-1 (P < 0.001) but not significant for FASLG (P = 0.06). A focused unsupervised hierarchal clustering analysis, including T-cell activation and immune checkpoint-related proteins, identified clusters of patients with higher CB rate and higher tumor expression of leukocyte or T-cell markers (CD3, CD45, granzyme B). Thirty-six proteins increased significantly during immunotherapy. Several proteins (including FASLG, checkpoint proteins, and immune activation markers) increased independently of response during immunotherapy but did not increase in the cohort of patients treated with chemotherapy. CONCLUSIONS Circulating levels of immune-related proteins like FASLG and Gal-1 might be used to predict the efficacy of checkpoint inhibitors in patients with metastatic PDAC.
Collapse
Affiliation(s)
- T D Christensen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev.
| | | | - S Theile
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev
| | - K Madsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev
| | - S C Lindgaard
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev
| | - J P Hasselby
- Department of Pathology, Copenhagen University Hospital-Rigshospitalet, Copenhagen
| | - D L Nielsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen
| | - J S Johansen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen; Department of Medicine, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - I M Chen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev
| |
Collapse
|
417
|
Xu ZJ, Li JA, Cao ZY, Xu HX, Ying Y, Xu ZH, Liu RJ, Guo Y, Zhang ZX, Wang WQ, Liu L. Construction of S100 family members prognosis prediction model and analysis of immune microenvironment landscape at single-cell level in pancreatic adenocarcinoma: a tumor marker prognostic study. Int J Surg 2024; 110:3591-3605. [PMID: 38498399 PMCID: PMC11175822 DOI: 10.1097/js9.0000000000001293] [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: 10/16/2023] [Accepted: 02/22/2024] [Indexed: 03/20/2024]
Abstract
Pancreatic adenocarcinoma characterized by a mere 10% 5-year survival rate, poses a formidable challenge due to its specific anatomical location, making tumor tissue acquisition difficult. This limitation underscores the critical need for novel biomarkers to stratify this patient population. Accordingly, this study aimed to construct a prognosis prediction model centered on S100 family members. Leveraging six S100 genes and their corresponding coefficients, an S100 score was calculated to predict survival outcomes. The present study provided comprehensive internal and external validation along with power evaluation results, substantiating the efficacy of the proposed model. Additionally, the study explored the S100-driven potential mechanisms underlying malignant progression. By comparing immune cell infiltration proportions in distinct patient groups with varying prognoses, the research identified differences driven by S100 expression. Furthermore, the analysis explored significant ligand-receptor pairs between malignant cells and immune cells influenced by S100 genes, uncovering crucial insights. Notably, the study identified a novel biomarker capable of predicting the sensitivity of neoadjuvant chemotherapy, offering promising avenues for further research and clinical application.
Collapse
Affiliation(s)
- Zi-jin Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
- Department of General Surgery, QingPu Branch of Zhongshan Hospital, Fudan University
| | - Jian-ang Li
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Ze-yuan Cao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, People’s Republic of China
| | - Hua-xiang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Ying Ying
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Zhi-hang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Run-jie Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Yuquan Guo
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Zi-xin Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Wen-quan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University
- Cancer Center, Zhongshan Hospital, Fudan University
- Department of Oncology, Shanghai Medical College, Fudan University
| |
Collapse
|
418
|
Qin Q, Yu R, Eriksson JE, Tsai HI, Zhu H. Cancer-associated fibroblasts in pancreatic ductal adenocarcinoma therapy: Challenges and opportunities. Cancer Lett 2024; 591:216859. [PMID: 38615928 DOI: 10.1016/j.canlet.2024.216859] [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: 10/25/2023] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a solid organ malignancy with a high mortality rate. Statistics indicate that its incidence has been increasing as well as the associated deaths. Most patients with PDAC show poor response to therapies making the clinical management of this cancer difficult. Stromal cells in the tumor microenvironment (TME) contribute to the development of resistance to therapy in PDAC cancer cells. Cancer-associated fibroblasts (CAFs), the most prevalent stromal cells in the TME, promote a desmoplastic response, produce extracellular matrix proteins and cytokines, and directly influence the biological behavior of cancer cells. These multifaceted effects make it difficult to eradicate tumor cells from the body. As a result, CAF-targeting synergistic therapeutic strategies have gained increasing attention in recent years. However, due to the substantial heterogeneity in CAF origin, definition, and function, as well as high plasticity, majority of the available CAF-targeting therapeutic approaches are not effective, and in some cases, they exacerbate disease progression. This review primarily elucidates on the effect of CAFs on therapeutic efficiency of various treatment modalities, including chemotherapy, radiotherapy, immunotherapy, and targeted therapy. Strategies for CAF targeting therapies are also discussed.
Collapse
Affiliation(s)
- Qin Qin
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China
| | - Rong Yu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China
| | - John E Eriksson
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, FI-20520 Finland
| | - Hsiang-I Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
419
|
Takeda T, Sasaki T, Okamoto T, Hirai T, Ishitsuka T, Yamada M, Nakagawa H, Furukawa T, Mie T, Kasuga A, Ozaka M, Sasahira N. Bone loss over time and risk of osteoporosis in advanced pancreatic cancer. Jpn J Clin Oncol 2024; 54:667-674. [PMID: 38452123 DOI: 10.1093/jjco/hyae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Pancreatic cancer has a high risk of developing osteoporosis. However, the impact of osteoporosis has not been well-studied. This study aimed to evaluate bone loss over time and risk of osteoporosis in patients with advanced pancreatic cancer. METHODS We retrospectively examined consecutive patients with unresectable pancreatic cancer who had evaluable computed tomography before treatment and at 1-year follow-up. Bone mineral density at the first lumbar vertebra was measured on computed tomography, and osteoporosis was defined as bone mineral density < 135 Hounsfield units. The prevalence and risk factors for osteoporosis, changes in bone mineral density over time and incidence of bone fractures were analyzed. RESULTS Three hundred eighty patients were included. Osteoporosis was associated with older age, female sex, low body mass index and poor performance status at baseline. A consistent decrease in bone mineral density was observed over time regardless of age, sex or disease status, resulting in an increase in the prevalence of osteoporosis over time (47% at baseline, 79% at 1 year, 88% at 2 years, 89% at 3 years, 95% at 4 years and 100% at 5 years). Changes in bone mineral density from baseline were greater in patients with locally-advanced pancreatic cancer, in those who received modified FOLFIRINOX or S-IROX for more than 3 months, and in those who received radiation therapy. Incident fractures developed in 45 patients (12%) during follow-up. CONCLUSIONS Osteoporosis and osteoporotic fractures were highly prevalent in patients with advanced pancreatic cancer. This study highlights the importance of screening for osteoporosis in such patients.
Collapse
Affiliation(s)
- Tsuyoshi Takeda
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takashi Sasaki
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takeshi Okamoto
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tatsuki Hirai
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takahiro Ishitsuka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Manabu Yamada
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroki Nakagawa
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takaaki Furukawa
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takafumi Mie
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akiyoshi Kasuga
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Naoki Sasahira
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
420
|
Yu S, Wang M, Zhang H, Guo X, Qin R. Resistance to gemcitabine is mediated by the circ_0036627/miR-145/S100A16 axis in pancreatic cancer. J Cell Mol Med 2024; 28:e18444. [PMID: 38924205 PMCID: PMC11196374 DOI: 10.1111/jcmm.18444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 04/27/2024] [Accepted: 05/11/2024] [Indexed: 06/28/2024] Open
Abstract
The development of gemcitabine (GEM) resistance severely limits the treatment efficacy in pancreatic cancer (PC) and increasing evidence highlights the vital roles of circular RNAs (circRNAs) in the tumorigenesis, progression and drug resistance of PC. However, the circRNAs underlying GEM resistance development of PC remains to be clarified. The current research aims to unveil the roles of circ_0036627 in dictating the aggressiveness and GEM sensitivity in PC. We reported the increased expression of circ_0036627 in PC tissues and PC cell lines. Elevated circ_0036627 expression level was correlated with advanced tumour grade and poor overall survival in PC patients. Functional assays and in vivo experiments demonstrated that circ_0036627 overexpression was required for the proliferation, migration invasion and GEM resistance in PC cells. circ_0036627 knockdown suppressed tumour development in vivo. The molecular analysis further showed that circ_0036627 increased S100A16 expression by sponging microRNA-145 (miR-145), a tumour-suppressive miRNA that could significantly attenuate PC cell proliferation, migration, invasion and GEM resistance. Furthermore, our findings suggested that S100A16 acted as an oncogenic factor to promote aggressiveness and GEM resistance in PC cells. In conclusion, the current findings provide new mechanistic insights into PC aggressiveness and GEM resistance, suggesting the critical role of circ_0036627/miR-145/S100A16 axis in PC progression and drug resistance development and offering novel therapeutic targets for PC therapy.
Collapse
Affiliation(s)
- Shuo Yu
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Min Wang
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Hang Zhang
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Xingjun Guo
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Renyi Qin
- Department of Biliary‐Pancreatic SurgeryAffiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
421
|
Su YY, Chiang NJ, Chiu TJ, Huang CJ, Hsu SJ, Lin HC, Yang SH, Yang Y, Chou WC, Chen YY, Bai LY, Li CP, Chen JS. Systemic treatments in pancreatic cancer: Taiwan pancreas society recommendation. Biomed J 2024; 47:100696. [PMID: 38169173 PMCID: PMC11332987 DOI: 10.1016/j.bj.2023.100696] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/05/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with a poor prognosis. Over the past decade, significant therapeutic advancements have improved the survival rates of patients with pancreatic cancer. One of the primary factors contributing to these positive outcomes is the evolution of chemotherapy, from monotherapy to doublet or triplet regimens, and the integration of multimodal approaches. Additionally, targeted agents tailored to patients with specific genetic alterations and the development of cell therapies show promise in benefiting certain subpopulations. This article focuses on examining pivotal studies that explore the role of chemotherapy in neoadjuvant, adjuvant, maintenance, and salvage settings; highlights interesting findings related to cell therapy; and provides an overview of ongoing trials concerning metastatic settings. This review primarily aimed to offer recommendations based on therapeutic evidence, recent advancements in new treatment combinations, and the most innovative approaches. A unique aspect of this review is the inclusion of published papers on clinical trials and real-world data in Taiwan, thus adding a valuable perspective to the overall analysis.
Collapse
Affiliation(s)
- Yung-Yeh Su
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Internal Medicine, Kaohsiung Medical University Hospital, and Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Nai-Jung Chiang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Therapeutic and Research Center of Pancreatic Cancer, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tai-Jan Chiu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chien-Jui Huang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shao-Jung Hsu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Therapeutic and Research Center of Pancreatic Cancer, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsin-Chen Lin
- Division of Medical Oncology, Department of Oncology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shih-Hung Yang
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Youngsen Yang
- Division of Cancer Prevention and Control, Department of Oncology, Taichung Veterans General Hospital, Taichung, Taiwan; College of Medicine, School of Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Chi Chou
- Chang Gung University College of Medicine, Taoyuan, Taiwan; Department of Hematology-Oncology, Linkou Chang Gung Memorial Hospital, Taiwan
| | - Yen-Yang Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Li-Yuan Bai
- College of Medicine, School of Medicine, China Medical University, Taichung, Taiwan; Division of Hematology and Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chung-Pin Li
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Therapeutic and Research Center of Pancreatic Cancer, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Clinical Skills Training, Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Jen-Shi Chen
- Chang Gung University College of Medicine, Taoyuan, Taiwan; Department of Hematology-Oncology, Linkou Chang Gung Memorial Hospital, Taiwan.
| |
Collapse
|
422
|
Belfiori G, Crippa S, Pagnanelli M, Gasparini G, Aleotti F, Camisa PR, Partelli S, Pecorelli N, De Stefano F, Schiavo Lena M, Palumbo D, Tamburrino D, Reni M, Falconi M. Very Early Recurrence After Curative Resection for Pancreatic Ductal Adenocarcinoma: Proof of Concept for a "Biological R2 Definition". Ann Surg Oncol 2024; 31:4084-4095. [PMID: 38459416 DOI: 10.1245/s10434-024-15105-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 02/12/2024] [Indexed: 03/10/2024]
Abstract
PURPOSE Very early recurrence after radical surgery for pancreatic ductal adenocarcinoma (PDAC) has been poorly investigated. This study was designed to evaluate this group of patients who developed recurrence, within 12 weeks after surgery, defined as "biological R2 resections (bR2)." METHODS Data from patients who underwent surgical resection as upfront procedure or after neoadjuvant treatment for PDAC between 2015 and 2019 were analyzed. Disease-free, disease-specific survival, and independent predictors of early recurrence were examined. The same analysis was performed separately for upfront and neoadjuvant treated patients. RESULTS Of the 573 patients included in the study, 63 (11%) were classified as bR2. The rate of neoadjuvant treatment was similar in bR2 and in the remaining patients (44 vs. 42%, p = 0.78). After a median follow-up of 27 months, median DFS and DSS for the entire cohort were 17 and 43 months, respectively. Median DSS of bR2 group was 13 months. The only preoperative identifiable independent predictor of very early recurrence was body-tail site lesion, whereas all other were pathological: higher pT (8th classification), G3 differentiation, and high lymph node ratio. These predictors were confirmed for patients undergoing upfront surgery, whereas in the neoadjuvant group the only independent predictor was pT. CONCLUSIONS One of ten patients with "radical" resected PDAC relapses very early after surgery (bR2); hence, imaging must be routinely repeated within 12 weeks. Despite higher biological aggressiveness and worse pathology, this bR2 cluster eludes our preoperative examinations.
Collapse
Affiliation(s)
- Giulio Belfiori
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Crippa
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita Salute San Raffaele, Milan, Italy
| | | | - Giulia Gasparini
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita Salute San Raffaele, Milan, Italy
| | - Francesca Aleotti
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Stefano Partelli
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita Salute San Raffaele, Milan, Italy
| | - Nicolò Pecorelli
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita Salute San Raffaele, Milan, Italy
| | - Federico De Stefano
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita Salute San Raffaele, Milan, Italy
| | - Marco Schiavo Lena
- Department of Pathology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Diego Palumbo
- Università Vita Salute San Raffaele, Milan, Italy
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Domenico Tamburrino
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Michele Reni
- Università Vita Salute San Raffaele, Milan, Italy
- Department of Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Falconi
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Università Vita Salute San Raffaele, Milan, Italy.
| |
Collapse
|
423
|
Wode K, Kienle GS, Björ O, Fransson P, Sharp L, Elander NO, Bernhardson BM, Johansson B, Edwinsdotter Ardnor C, Scheibling U, Hök Nordberg J, Henriksson R. Mistletoe Extract in Patients With Advanced Pancreatic Cancer: a Double-Blind, Randomized, Placebo-Controlled Tial (MISTRAL). DEUTSCHES ARZTEBLATT INTERNATIONAL 2024; 121:347-354. [PMID: 38915151 PMCID: PMC11539882 DOI: 10.3238/arztebl.m2024.0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND Patients with advanced pancreatic cancer have limited survival and few treatment options. We studied whether mistletoe extract (ME), in addition to comprehensive oncological treatment and palliative care, prolongs overall survival (OS) and improves health-related quality of life (HRQoL). METHODS The double-blind, placebo-controlled MISTRAL trial was conducted in Swedish oncology centers. The main inclusion criteria were advanced exocrine pancreatic cancer and Eastern Cooperative Oncology Group (ECOG) performance status 0-2. The subjects were randomly assigned to ME (n=143) or placebo (n=147) and were stratified by study site and by eligibility (yes/no) for palliative chemotherapy (June 2016-December 2021). ME or placebo was injected subcutaneously three times a week for nine months. The primary endpoint was overall survival (OS); one of the secondary endpoints was the HRQoL dimension global health/QoL (EORTC-QLQ-C30), as assessed at seven time points over nine months. Trial registration: EudraCT 2014-004552-64, NCT02948309. RESULTS No statistically significant benefit of adding ME to standard treatment was seen with respect to either OS or global health/ QoL. The adjusted hazard ratio for OS was 1.13 [0.89; 1.44], with a median survival time of 7.8 and 8.3 months for ME and placebo, respectively. The figures for the HRQoL dimension "global health/QoL" were similar in the two groups (p=0.86). The number, severity, and outcome of the reported adverse events were similar as well, except for more common local skin reactions at ME injection sites (66% vs. 1%). CONCLUSION ME is unlikely to have a clinically significant effect on OS or the HRQoL dimension global health/QoL when administered in patients with advanced pancreatic cancer in addition to comprehensive cancer care.
Collapse
Affiliation(s)
- Kathrin Wode
- Department of Radiation Sciences/Oncology, Umeå University, Umeå, Sweden
- Department of Neurobiology, Caring Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Regional Cancer Centre Stockholm Gotland, Stockholm, Sweden
| | - Gunver Sophia Kienle
- Center for Complementary Medicine, Department of Medicine II, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute for Applied Epistemology and Medical Methodology at Witten/Herdecke University (IFAEMM), Freiburg, Germany
| | - Ove Björ
- Department of Radiation Sciences/Oncology, Umeå University, Umeå, Sweden
| | - Per Fransson
- Department of Nursing, Umeå University, Umeå, Sweden
| | - Lena Sharp
- Regional Cancer Centre Stockholm Gotland, Stockholm, Sweden
- Department of Nursing, Umeå University, Umeå, Sweden
| | - Nils O. Elander
- Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Britt-Marie Bernhardson
- Department of Neurobiology, Caring Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Birgit Johansson
- Department of Oncology, Västmanlands Hospital, Västerås, Schweden
| | | | - Ursula Scheibling
- Department of Oncology, Ryhov County Hospital, Jönköping, Schweden
- Palliative Care Unit Västerås, Schweden
| | - Johanna Hök Nordberg
- Department of Neurobiology, Caring Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Regional Cancer Centre Stockholm Gotland, Stockholm, Sweden
| | - Roger Henriksson
- Department of Radiation Sciences/Oncology, Umeå University, Umeå, Sweden
| |
Collapse
|
424
|
Kodama T, Imajima T, Shimokawa M, Otsuka T, Kawahira M, Nakazawa J, Hori T, Shibuki T, Arima S, Ido A, Miwa K, Okabe Y, Koga F, Ueda Y, Kubotsu Y, Shimokawa H, Takeshita S, Nishikawa K, Komori A, Otsu S, Hosokawa A, Sakai T, Sakai K, Oda H, Kawahira M, Arita S, Honda T, Taguchi H, Tsuneyoshi K, Kawaguchi Y, Fujita T, Sakae T, Shirakawa T, Mizuta T, Mitsugi K. A multicenter retrospective observational NAPOLEON2 study of nanoliposomal irinotecan with fluorouracil and folinic acid in patients with unresectable pancreatic cancer. Sci Rep 2024; 14:12422. [PMID: 38816500 PMCID: PMC11139902 DOI: 10.1038/s41598-024-63172-y] [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: 02/24/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024] Open
Abstract
Nanoliposomal irinotecan with fluorouracil and folinic acid (NFF) is a standard regimen after gemcitabine-based therapy for patients with unresectable or recurrent pancreatic cancer. However, there are limited clinical data on its efficacy and safety in the real-world. We therefore initiated a retrospective and prospective observational study (NAPOLEON-2). The results of the retrospective part were reported herein. In this retrospective study, we evaluated 161 consecutive patients who received NFF as second-or-later-line regimen. The main endpoint was overall survival (OS), and the other endpoints were response rate, disease control rate, progression-free survival (PFS), dose intensity, and adverse events (AEs). The median age was 67 years (range, 38-85 years). The median OS and PFS were 8.1 and 3.4 months, respectively. The objective response and disease control rates were 5% and 52%, respectively. The median relative dose intensity was 81.6% for nanoliposomal irinotecan and 82.9% for fluorouracil. Grade 3 or 4 hematological and nonhematological AEs occurred in 47 and 42 patients, respectively. Common grade 3 or 4 AEs included neutropenia (24%), anorexia (12%), and leukocytopenia (12%). Subanalysis of patients treated with second-line and third-or-later-line demonstrated no statistical significant difference in OS (7.6 months vs. 9.1 months, respectively; hazard ratio, 0.92; 95% confidence interval, 0.64-1.35; p = 0.68). In conclusion, NFF has acceptable efficacy and safety profile even in real-world clinical settings. The prospective study is in progress to validate these findings.
Collapse
Affiliation(s)
- Tomoko Kodama
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-Cho, Kagoshima-Shi, Kagoshima, 890-8760, Japan
| | - Takashi Imajima
- Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-Cho, Sasebo-Shi, Nagasaki, 857-8575, Japan
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi‑ku, Fukuoka-Shi, Fukuoka, 812-8582, 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 Internal Medicine, Minato Medical Clinic, 3-11-3 Nagahama, Chuo-Ku, Fukuoka-Shi, Fukuoka, 810-0072, Japan
| | - Masahiro Kawahira
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-Cho, Kagoshima-Shi, Kagoshima, 890-8760, Japan
| | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-Cho, Kagoshima-Shi, Kagoshima, 890-8760, Japan
| | - Takeshi Hori
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-Cho, Kagoshima-Shi, Kagoshima, 890-8760, Japan
| | - Taro Shibuki
- Department for the Promotion of Drug and Diagnostic Development, Division of Drug and Diagnostic Development Promotion, Translational Research Support Office, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa-Shi, Chiba, 277-8577, Japan
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa-Shi, Chiba, 277-8577, 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
| | - Akio Ido
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima-Shi, Kagoshima, 890-8520, Japan
| | - Keisuke Miwa
- Multidisciplinary Treatment Cancer Center, Kurume University Hospital, 67 Asahi-Machi, Kurume-Shi, Fukuoka, 830-0011, Japan
| | - Yoshinobu Okabe
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume-Shi, Fukuoka, 830-0011, 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
| | - Yoshihito Kubotsu
- Department of Internal Medicine, Karatsu Red Cross Hospital, 2430 Watada, Karatsu-Shi, Saga, 847-8588, Japan
| | - Hozumi Shimokawa
- Department of Hematology and Oncology, Japan Community Healthcare Organization Kyushu Hospital, 1-8-1 Kishinoura, Yahatanishi-Ku, Kitakyushu-Shi, Fukuoka, 806-8501, Japan
| | - Shigeyuki Takeshita
- Department of Gastroenterology, Japanese Red Cross Nagasaki Genbaku Hospital, 3-15 Morimachi, Nagasaki-Shi, Nagasaki, 852-8511, Japan
| | - Kazuo Nishikawa
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-Machi, Yufu-Shi, Oita, 879-5593, Japan
| | - Azusa Komori
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-Machi, Yufu-Shi, Oita, 879-5593, Japan
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, 160 Kou, Minamiumemoto-Machi, Matsuyama-Shi, Ehime, 791-0280, Japan
| | - Satoshi Otsu
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-Machi, Yufu-Shi, Oita, 879-5593, Japan
| | - Ayumu Hosokawa
- Department of Clinical Oncology, University of Miyazaki Hospital, 5200 Kiyotakechoukihara, Miyazaki-Shi, Miyazaki, 889-1692, Japan
| | - Tatsunori Sakai
- Department of Medical Oncology, National Hospital Organization Kumamoto Medical Center, 1-5 Ninomaru, Chuo-Ku, Kumamoto-Shi, Kumamoto, 860-0008, Japan
| | - Kenji Sakai
- Department of Medical Oncology, National Hospital Organization Kumamoto Medical Center, 1-5 Ninomaru, Chuo-Ku, Kumamoto-Shi, Kumamoto, 860-0008, Japan
- Department of Clinical Oncology, Japan Community Health Care Organization Hitoyoshi Medical Center, 35 Oikamimachi, Hitoyoshi-Shi, Kumamoto, 868-8555, Japan
| | - Hisanobu Oda
- Division of Integrative Medical Oncology, Saiseikai Kumamoto Hospital, 5-3-1 Chikami, Minami-Ku, Kumamoto-Shi, Kumamoto, 861-4193, Japan
| | - Machiko Kawahira
- Department of Gastroenterology, Kagoshima Kouseiren Hospital, 1-13-1 Yojirou, Kagoshima-Shi, Kagoshima, 890-0062, Japan
| | - Shuji Arita
- Department of Chemotherapy, Miyazaki Prefectural Miyazaki Hospital, 5-30 Kita-Takamatsucho, Miyazaki, 880-8510, 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
| | - Hiroki Taguchi
- Department of Gastroenterology, Izumi General Medical Center, 520 Myoujin-Cho, Izumi-Shi, Kagoshima, 899-0131, Japan
- Department of Gastroenterology, Kagoshima City Hospital, 37-1 Uearata-Cho, Kagoshima-Shi, Kagoshima, 890-8760, Japan
| | - Kengo Tsuneyoshi
- Department of Gastroenterology, Izumi General Medical Center, 520 Myoujin-Cho, Izumi-Shi, Kagoshima, 899-0131, Japan
| | - Yasunori Kawaguchi
- Department of Gastroenterology, Asakura Medical Association Hospital, 422-1 Raiha, Asakura-Shi, Fukuoka, 838-0069, Japan
| | - Toshihiro Fujita
- Department of Gastroenterology, Saiseikai Sendai Hospital, 2-46 Harada-Cho, Satsumasendai-Shi, Kagoshima, 895-0074, Japan
| | - Takahiro Sakae
- Department of Gastroenterology, Saiseikai Sendai Hospital, 2-46 Harada-Cho, Satsumasendai-Shi, Kagoshima, 895-0074, Japan
| | - Tsuyoshi Shirakawa
- Department of Medical Checkup Center, Eikoh Hospital, 3-8-15 Befu-Nishi, Shime-Machi, Kasuya-Gun, Fukuoka, 811-2232, Japan.
- Clinical Hematology Oncology Treatment Study Group, 1-14-6 Muromi-Gaoka, Nishi-Ku, Fukuoka-Shi, Fukuoka, 819-0030, Japan.
| | - Toshihiko Mizuta
- 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
| |
Collapse
|
425
|
Jang DK, Kim YA, Lee JW, Kim HJ, Lee YS, Chun JW, Lee JC, Woo SM, Hwang JH. The Trends and Outcomes of Initial Palliative Chemotherapy in Patients with Pancreatic Cancer in Korea Based on National Health Insurance Service Data. J Clin Med 2024; 13:3229. [PMID: 38892939 PMCID: PMC11172641 DOI: 10.3390/jcm13113229] [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: 04/22/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Background/Objectives: The survival rate of patients with pancreatic cancer (PC) has improved gradually since the introduction of FOLFIRINOX (FFX) and gemcitabine + albumin-bound paclitaxel (GnP) regimens. However, the trends and outcomes of initial palliative chemotherapy before and after the advent of these regimens and their contribution to survival rates are not well understood. This study aimed to investigate this in patients with PC in Korea using claims data from the National Health Insurance Service (NHIS). Methods: Patients diagnosed with PC who underwent initial palliative chemotherapy between 2007 and 2019 were identified from the NHIS database. Patient demographics, comorbidities, chemotherapy regimens, and survival rates were analyzed using follow-up data up to 2020. Results: In total, 14,760 patients (mean age, 63.78 ± 10.18 years; men, 59.19%) were enrolled. As initial palliative chemotherapy, 3823 patients (25.90%) received gemcitabine alone; 2779 (18.83%) received gemcitabine + erlotinib; 1948 (13.20%) received FFX; and 1767 (11.97%) received GnP. The median survival values were 15.00 months for FFX; 11.04 months for GnP; 8.40 months for gemcitabine alone; and 8.51 months for gemcitabine + erlotinib. The adjusted hazard ratio (aHR) for GnP vs. FFX was 1.291 (95% CI, 1.206-1.383) in the multivariate Cox regression analysis of mortality. Radiation therapy (aHR, 0.667; 95% CI, 0.612-0.728) and second-line chemotherapy (aHR, 0.639; 95% CI, 0.597-0.684) were significantly associated with improved survival. Conclusions: Our study found that first-line chemotherapy with FFX was associated with significantly longer survival than the other regimens, although caution is needed in interpreting the results.
Collapse
Affiliation(s)
- Dong Kee Jang
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Republic of Korea;
| | - Young Ae Kim
- Division of Cancer Control & Policy, National Cancer Control Institute, National Cancer Center, Goyang 10408, Republic of Korea; (Y.A.K.); (J.W.L.); (H.-J.K.)
| | - Jang Won Lee
- Division of Cancer Control & Policy, National Cancer Control Institute, National Cancer Center, Goyang 10408, Republic of Korea; (Y.A.K.); (J.W.L.); (H.-J.K.)
| | - Hak-June Kim
- Division of Cancer Control & Policy, National Cancer Control Institute, National Cancer Center, Goyang 10408, Republic of Korea; (Y.A.K.); (J.W.L.); (H.-J.K.)
| | - Yoon Suk Lee
- Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang 10380, Republic of Korea;
| | - Jung Won Chun
- Research Institute, Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea;
| | - Jong-Chan Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| | - Sang Myung Woo
- Research Institute, Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang 10408, Republic of Korea;
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| |
Collapse
|
426
|
Machover D, Almohamad W, Castagné V, Desterke C, Gomez L, Goldschmidt E. Treatment of patients with carcinomas in advanced stages with 5-fluorouracil, folinic acid and pyridoxine in tandem. Sci Rep 2024; 14:12054. [PMID: 38802419 PMCID: PMC11130240 DOI: 10.1038/s41598-024-62860-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/22/2024] [Indexed: 05/29/2024] Open
Abstract
The effect of high-dose pyridoxine (PN) on activity of 5-fluorouracil (FUra) and folinic acid (FA)-containing regimens was studied in 50 patients including 14 with digestive tract, and 36 with breast carcinomas (BC) in advanced stages with poor prognostic characteristics. Patients with colorectal, and pancreas adenocarcinoma received oxaliplatin, irinotecan, FUra, FA (Folfirinox), and patients with squamous cell carcinoma of the esophagus had paclitaxel, carboplatin, FUra, FA (TCbF). Patients with BC received AVCF (doxorubicin, vinorelbine, cyclophosphamide, FUra, FA) followed by TCbF or TCbF only, and patients who overexpressed HER2 received TCbF plus trastuzumab and pertuzumab. PN (1000-3000 mg/day iv) preceded each administration of FUra and FA. 47 patients (94%) responded, including 16 (32%) with CR. Median tumor reduction was 93%. Median event-free survival (EFS) was 37.7 months. The 25 patients with tumor shrinkage ≥ 91% had EFS of 52% from 42 months onwards. Unexpected toxicity did not occur. PN enhances potency of chemotherapy regimens comprising FUra and FA.
Collapse
Affiliation(s)
- David Machover
- INSERM U935-UA09, University Paris-Saclay, Paul-Brousse Hospital, 12, Avenue Paul-Vaillant-Couturier, 94800, Villejuif, France.
| | - Wathek Almohamad
- Department of Medical Oncology, University Paris-Saclay, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris (APHP), 94800, Villejuif, France
| | - Vincent Castagné
- Department of Pharmacy, University Paris-Saclay, Paul-Brousse Hospital, APHP, 94800, Villejuif, France
| | - Christophe Desterke
- INSERM U935-UA09, University Paris-Saclay, Paul-Brousse Hospital, 12, Avenue Paul-Vaillant-Couturier, 94800, Villejuif, France
| | - Léa Gomez
- Department of Biophysics and Nuclear Medicine, University Paris-Saclay, Kremlin-Bicêtre Hospital, APHP, 94270, Le Kremlin-Bicêtre, France
| | - Emma Goldschmidt
- Department of Medical Oncology, University Paris-Saclay, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris (APHP), 94800, Villejuif, France
| |
Collapse
|
427
|
Ma Y, Dumesny C, Dong L, Ang CS, Asadi K, Zhan Y, Nikfarjam M, He H. Inhibition of P21-activated kinases 1 and 4 synergistically suppresses the growth of pancreatic cancer by stimulating anti-tumour immunity. Cell Commun Signal 2024; 22:287. [PMID: 38797819 PMCID: PMC11129409 DOI: 10.1186/s12964-024-01670-2] [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: 02/21/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal types of cancer, and KRAS oncogene occurs in over 90% of cases. P21-activated kinases (PAK), containing six members (PAK1 to 6), function downstream of KRAS. PAK1 and PAK4 play important roles in carcinogenesis, but their combinational effect remains unknown. In this study, we have determined the effect of dual inhibition of PAK1 and PAK4 in PDA progression using knockout (KO) cancer cell lines. METHODS Murine wild-type (WT) and PAK1KO pancreatic cancer cell lines were isolated from PAK1+/+ and PAK1-/- KPC (LSL-KrasG12D/+; LSL-Trp53 R172H/+; Pdx-1-Cre) mice. KPC PAK4KO and KPC PAK1&4 KO cell lines were generated from KPC WT and KPC PAK1KO cell lines respectively using the CRISPR-CAS9 gene knockout technique. PAK WT and KO cell lines were used in mouse models of pancreatic tumours. Cells and tumour tissue were also used in flow cytometry and proteomic studies. A human PDA tissue microarray was stained by immunohistochemistry. RESULTS Double knock out of PAK1 and PAK4 caused complete regression of tumour in a syngeneic mouse model. PAK4KO inhibited tumour growth by stimulating a rapid increase of cytotoxic CD8+ T cell infiltration. PAK1KO synergistically with PAK4KO increased cytotoxic CD8+ T cell infiltration and stimulated a sustained infiltration of CD8+ T cells at a later phase to overcome the immune evasion in the PAK4KO tumour. The human PDA tissue microarray study showed the important role of PAK1 and PAK4 in intra-tumoral T-cell function. CONCLUSION Our results demonstrated that dual inhibition of PAK1 and PAK4 synergistically suppressed PDA progression by stimulating cytotoxic CD8 + T cell response.
Collapse
Affiliation(s)
- Yi Ma
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
- Department of General Surgery, Monash Health, Clayton, VIC, Australia
| | - Chelsea Dumesny
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
| | - Li Dong
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
| | - Ching-Seng Ang
- Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Khashayar Asadi
- Department of Anatomical Pathology, Austin Health, Heidelberg, VIC, Australia
| | - Yifan Zhan
- Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Mehrdad Nikfarjam
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
- Department of Hepato-Pancreato-Biliary Surgery, Austin Health, Heidelberg, VIC, Australia
| | - Hong He
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia.
| |
Collapse
|
428
|
van Eijck CWF, Real FX, Malats N, Vadgama D, van den Bosch TPP, Doukas M, van Eijck CHJ, Mustafa DAM. GATA6 identifies an immune-enriched phenotype linked to favorable outcomes in patients with pancreatic cancer undergoing upfront surgery. Cell Rep Med 2024; 5:101557. [PMID: 38733987 PMCID: PMC11148804 DOI: 10.1016/j.xcrm.2024.101557] [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: 12/19/2023] [Revised: 02/14/2024] [Accepted: 04/16/2024] [Indexed: 05/13/2024]
Abstract
This study underscores GATA6's role in distinguishing classical and basal-like pancreatic ductal adenocarcinoma (PDAC) phenotypes. Retrospective studies associate GATA6 immunohistochemistry (IHC) expression with survival outcomes, warranting prospective validation. In a prospective treatment-naive cohort of patients with resected PDAC, GATA6 IHC proves a prognostic discriminator, associating high GATA6 expression with extended survival and the classical PDAC phenotype. However, GATA6's prognostic significance is numerically lower after gemcitabine-based neoadjuvant chemoradiotherapy compared to its significance in patients treated with upfront surgery. Furthermore, GATA6 is implicated in immunomodulation, although a comprehensive investigation of its immunological role is lacking. Treatment-naive PDAC tumors with varying GATA6 expression yield distinct immunological landscapes. Tumors highly expressing GATA6 show reduced infiltration of immunosuppressive regulatory T cells and M2 macrophages but increased infiltration of immune-stimulating, antigen-presenting, and activated T cells. Our findings caution against solely relying on GATA6 for molecular subtyping in clinical trials and open avenues for exploring immune-based combination therapies.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, Madrid, Spain.
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre, Madrid, Spain; Centro de Investigación Biomédica en Red-Cáncer, Madrid, Spain; Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, Madrid, Spain; Centro de Investigación Biomédica en Red-Cáncer, Madrid, Spain
| | - Disha Vadgama
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Thierry P P van den Bosch
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Michail Doukas
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Dana A M Mustafa
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Centre, Rotterdam, the Netherlands; The Tumor Immuno-Pathology Laboratory, Erasmus University Medical Centre, Rotterdam, the Netherlands.
| |
Collapse
|
429
|
Huai M, Wang Y, Li J, Pan J, Sun F, Zhang F, Zhang Y, Xu L. Intelligent nanovesicle for remodeling tumor microenvironment and circulating tumor chemoimmunotherapy amplification. J Nanobiotechnology 2024; 22:257. [PMID: 38755645 PMCID: PMC11097415 DOI: 10.1186/s12951-024-02467-8] [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: 12/13/2023] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
Imperceptible examination and unideal treatment effect are still intractable difficulties for the clinical treatment of pancreatic ductal adenocarcinoma (PDAC). At present, despite 5-fluorouracil (5-FU), as a clinical first-line FOLFIRINOX chemo-drug, has achieved significant therapeutic effects. Nevertheless, these unavoidable factors such as low solubility, lack of biological specificity and easy to induce immunosuppressive surroundings formation, severely limit their treatment in PDAC. As an important source of energy for many tumor cells, tryptophan (Trp), is easily degraded to kynurenine (Kyn) by indolamine 2,3- dioxygenase 1 (IDO1), which activates the axis of Kyn-AHR to form special suppressive immune microenvironment that promotes tumor growth and metastasis. However, our research findings that 5-FU can induce effectively immunogenic cell death (ICD) to further treat tumor by activating immune systems, while the secretion of interferon-γ (IFN-γ) re-induce the Kyn-AHR axis activation, leading to poor treatment efficiency. Therefore, a metal matrix protease-2 (MMP-2) and endogenous GSH dual-responsive liposomal-based nanovesicle, co-loading with 5-FU (anti-cancer drug) and NLG919 (IDO1 inhibitor), was constructed (named as ENP919@5-FU). The multifunctional ENP919@5-FU can effectively reshape the tumor immunosuppression microenvironment to enhance the effect of chemoimmunotherapy, thereby effectively inhibiting cancer growth. Mechanistically, PDAC with high expression of MMP-2 will propel the as-prepared nanovesicle to dwell in tumor region via shedding PEG on the nanovesicle surface, effectively enhancing tumor uptake. Subsequently, the S-S bond containing nanovesicle was cut via high endogenous GSH, leading to the continued release of 5-FU and NLG919, thereby enabling circulating chemoimmunotherapy to effectively cause tumor ablation. Moreover, the combination of ENP919@5-FU and PD-L1 antibody (αPD-L1) showed a synergistic anti-tumor effect on the PDAC model with abdominal cavity metastasis. Collectively, ENP919@5-FU nanovesicle, as a PDAC treatment strategy, showed excellent antitumor efficacy by remodeling tumor microenvironment to circulate tumor chemoimmunotherapy amplification, which has promising potential in a precision medicine approach.
Collapse
Affiliation(s)
- Manxiu Huai
- Department of Gastroenterology Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Yingjie Wang
- Nano Science and Technology Institute, University of Science and Technology of China, Suzhou, 215123, China
| | - Junhao Li
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai, 200433, China
| | - Jiaxing Pan
- Department of Gastroenterology Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Fang Sun
- Department of Gastroenterology Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Feiyu Zhang
- Department of Gastroenterology Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Yi Zhang
- Department of Gastroenterology Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| | - Leiming Xu
- Department of Gastroenterology Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
430
|
Guo W, Wu D, Xu J, Wang L. Editorial: Mechanism and treatment for pancreatic cancer metastases. Front Oncol 2024; 14:1424817. [PMID: 38807766 PMCID: PMC11130483 DOI: 10.3389/fonc.2024.1424817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 05/03/2024] [Indexed: 05/30/2024] Open
Affiliation(s)
- Wenyi Guo
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Dong Wu
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
| | - Jianwei Xu
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Lei Wang
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
431
|
Desai A, Xiao AH, Choi D, Toruner MD, Walden D, Halfdanarson TR, Alberts S, McWilliams RR, Mahipal A, Ahn D, Babiker H, Stybayeva G, Revzin A, Kizilbash S, Adjei A, Bekaii-Saab T, Mansfield AS, Carr RM, Ma WW. Molecular Characterization and Therapeutic Opportunities in KRAS Wildtype Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:1861. [PMID: 38791940 PMCID: PMC11119482 DOI: 10.3390/cancers16101861] [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: 03/30/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
PURPOSE To investigate the molecular characteristics of and potential for precision medicine in KRAS wildtype pancreatic ductal adenocarcinoma (PDAC). PATIENTS AND METHODS We investigated 27 patients with KRASWT PDAC at our institution. Clinical data were obtained via chart review. Tumor specimens for each subject were interrogated for somatic single nucleotide variants, insertion and deletions, and copy number variants by DNA sequencing. Gene fusions were detected from RNA-seq. A patient-derived organoid (PDO) was developed from a patient with a MET translocation and expanded ex vivo to predict therapeutic sensitivity prior to enrollment in a phase 2 clinical trial. RESULTS Transcriptomic analysis showed our cohort may be stratified by the relative gene expression of the KRAS signaling cascade. The PDO derived from our patient harboring a TFG-MET rearrangement was found to have in vitro sensitivity to the multi-tyrosine kinase inhibitor crizotinib. The patient was enrolled in the phase 2 SPARTA clinical trial and received monotherapy with vebrelitinib, a c-MET inhibitor, and achieved a partial and durable response. CONCLUSIONS KRASWT PDAC is molecularly distinct from KRASMUT and enriched with potentially actionable genetic variants. In our study, transcriptomic profiling revealed that the KRAS signaling cascade may play a key role in KRASWT PDAC. Our report of a KRASWT PDAC patient with TFG-MET rearrangement who responded to a cMET inhibitor further supports the pursuit of precision oncology in this sub-population. Identification of targetable mutations, perhaps through approaches like RNA-seq, can help enable precision-driven approaches to select optimal treatment based on tumor characteristics.
Collapse
Affiliation(s)
- Aakash Desai
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | | | - Daheui Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA; (D.C.); (G.S.); (A.R.)
| | - Merih D. Toruner
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Daniel Walden
- Division of Hematology and Oncology, Mayo Clinic, Phoenix, AZ 85054, USA; (D.W.); (D.A.); (T.B.-S.)
| | - Thorvardur R. Halfdanarson
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Steven Alberts
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Robert R. McWilliams
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Amit Mahipal
- Department of Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA;
| | - Daniel Ahn
- Division of Hematology and Oncology, Mayo Clinic, Phoenix, AZ 85054, USA; (D.W.); (D.A.); (T.B.-S.)
| | - Hani Babiker
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA; (D.C.); (G.S.); (A.R.)
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA; (D.C.); (G.S.); (A.R.)
| | - Sani Kizilbash
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Alex Adjei
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Tanios Bekaii-Saab
- Division of Hematology and Oncology, Mayo Clinic, Phoenix, AZ 85054, USA; (D.W.); (D.A.); (T.B.-S.)
| | - Aaron S. Mansfield
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Ryan M. Carr
- Department of Oncology, Mayo Clinic, Rochester, MN 55902, USA; (A.D.); (M.D.T.); (T.R.H.); (S.A.); (R.R.M.); (S.K.)
| | - Wen Wee Ma
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| |
Collapse
|
432
|
Long SA, Amparo AM, Goodhart G, Ahmad SA, Waters AM. Evaluation of KRAS inhibitor-directed therapies for pancreatic cancer treatment. Front Oncol 2024; 14:1402128. [PMID: 38800401 PMCID: PMC11116577 DOI: 10.3389/fonc.2024.1402128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Despite significant advancements in the treatment of other cancers, pancreatic ductal adenocarcinoma (PDAC) remains one of the world's deadliest cancers. More than 90% of PDAC patients harbor a Kirsten rat sarcoma (KRAS) gene mutation. Although the clinical potential of anti-KRAS therapies has long been realized, all initial efforts to target KRAS were unsuccessful. However, with the recent development of a new generation of KRAS-targeting drugs, multiple KRAS-targeted treatment options for patients with PDAC have entered clinical trials. In this review, we provide an overview of current standard of care treatment, describe RAS signaling and the relevance of KRAS mutations, and discuss RAS isoform- and mutation-specific differences. We also evaluate the clinical efficacy and safety of mutation-selective and multi-selective inhibitors, in the context of PDAC. We then provide a comparison of clinically relevant KRAS inhibitors to second-line PDAC treatment options. Finally, we discuss putative resistance mechanisms that may limit the clinical effectiveness of KRAS-targeted therapies and provide a brief overview of promising therapeutic approaches in development that are focused on mitigating these resistance mechanisms.
Collapse
Affiliation(s)
- Szu-Aun Long
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Amber M. Amparo
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Grace Goodhart
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Syed A. Ahmad
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Andrew M. Waters
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
433
|
Klein M, Warschkow R, Ukegjini K, Krstic D, Burri P, Chatziisaak D, Steffen T, Schmied B, Probst P, Tarantino I. The influence of delayed gastric emptying on quality of life after partial duodenopancreatectomy. Langenbecks Arch Surg 2024; 409:155. [PMID: 38727871 DOI: 10.1007/s00423-024-03345-5] [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: 02/21/2024] [Accepted: 05/05/2024] [Indexed: 05/15/2024]
Abstract
PURPOSE Quality of life (QoL) is temporarily compromised after pancreatic surgery, but no evidence for a negative impact of postoperative complications on QoL has been provided thus far. Delayed gastric emptying (DGE) is one of the most common complications after pancreatic surgery and is associated with a high level of distress. Therefore, the aim of this study was to analyse the influence of DGE on QoL. METHODS This single-centre retrospective study analysed QoL after partial duodenopancreatectomy (PD) via the European Organization for Research and Treatment of Cancer core questionnaire (QLQ-C30). The QoL of patients with and without postoperative DGE was compared. RESULTS Between 2010 and 2022, 251 patients were included, 85 of whom developed DGE (34%). Within the first postoperative year, compared to patients without DGE, those with DGE had a significantly reduced QoL, by 9.0 points (95% CI: -13.0 to -5.1, p < 0.001). Specifically, physical and psychosocial functioning (p = 0.020) decreased significantly, and patients with DGE suffered significantly more from fatigue (p = 0.010) and appetite loss (p = 0.017) than patients without DGE. After the first postoperative year, there were no significant differences in QoL or symptom scores between patients with DGE and those without DGE. CONCLUSION Patients who developed DGE reported a significantly reduced QoL and reduced physical and psychosocial functioning within the first year after partial pancreatoduodenectomy compared to patients without DGE.
Collapse
Affiliation(s)
- Marie Klein
- Department of General, Visceral, Endocrine, and Transplant Surgery, Cantonal Hospital of St. Gallen, Rorschacherstrasse 95, St. Gallen, CH-9007, Switzerland.
| | - Rene Warschkow
- Department of General, Visceral, Endocrine, and Transplant Surgery, Cantonal Hospital of St. Gallen, Rorschacherstrasse 95, St. Gallen, CH-9007, Switzerland
| | - Kristjan Ukegjini
- Department of General, Visceral, Endocrine, and Transplant Surgery, Cantonal Hospital of St. Gallen, Rorschacherstrasse 95, St. Gallen, CH-9007, Switzerland
| | - Daniel Krstic
- Department of General, Visceral, Endocrine, and Transplant Surgery, Cantonal Hospital of St. Gallen, Rorschacherstrasse 95, St. Gallen, CH-9007, Switzerland
| | - Pascal Burri
- Department of General, Visceral, Endocrine, and Transplant Surgery, Cantonal Hospital of St. Gallen, Rorschacherstrasse 95, St. Gallen, CH-9007, Switzerland
| | - Dimitrios Chatziisaak
- Department of General, Visceral, Endocrine, and Transplant Surgery, Cantonal Hospital of St. Gallen, Rorschacherstrasse 95, St. Gallen, CH-9007, Switzerland
| | - Thomas Steffen
- Department of General, Visceral, Endocrine, and Transplant Surgery, Cantonal Hospital of St. Gallen, Rorschacherstrasse 95, St. Gallen, CH-9007, Switzerland
| | - Bruno Schmied
- Department of General, Visceral, Endocrine, and Transplant Surgery, Cantonal Hospital of St. Gallen, Rorschacherstrasse 95, St. Gallen, CH-9007, Switzerland
| | - Pascal Probst
- Department of Surgery, Cantonal Hospital Thurgau, Frauenfeld, Switzerland
| | - Ignazio Tarantino
- Department of General, Visceral, Endocrine, and Transplant Surgery, Cantonal Hospital of St. Gallen, Rorschacherstrasse 95, St. Gallen, CH-9007, Switzerland
| |
Collapse
|
434
|
Jamali M, Barar E, Shi J. Unveiling the Molecular Landscape of Pancreatic Ductal Adenocarcinoma: Insights into the Role of the COMPASS-like Complex. Int J Mol Sci 2024; 25:5069. [PMID: 38791111 PMCID: PMC11121229 DOI: 10.3390/ijms25105069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is poised to become the second leading cause of cancer-related death by 2030, necessitating innovative therapeutic strategies. Genetic and epigenetic alterations, including those involving the COMPASS-like complex genes, have emerged as critical drivers of PDAC progression. This review explores the genetic and epigenetic landscape of PDAC, focusing on the role of the COMPASS-like complex in regulating chromatin accessibility and gene expression. Specifically, we delve into the functions of key components such as KDM6A, KMT2D, KMT2C, KMT2A, and KMT2B, highlighting their significance as potential therapeutic targets. Furthermore, we discuss the implications of these findings for developing novel treatment modalities for PDAC.
Collapse
Affiliation(s)
- Marzieh Jamali
- Department of Pathology & Clinical Labs, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Erfaneh Barar
- Liver and Pancreatobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Jiaqi Shi
- Department of Pathology & Clinical Labs, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
435
|
Shinde A, Panchal K, Patra P, Singh S, Enakolla S, Paliwal R, Chaurasiya A. QbD Enabled Development and Evaluation of Pazopanib Loaded Nanoliposomes for PDAC Treatment. AAPS PharmSciTech 2024; 25:97. [PMID: 38710894 DOI: 10.1208/s12249-024-02806-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: 02/09/2024] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the highly fatal types of cancer with high mortality/incidence. Considering the crucial role of vascular endothelial growth factor (VEGF) in PDAC progression, its inhibition can be a viable strategy for the treatment. Pazopanib, a second-generation VEGF inhibitor, is approved for the treatment of various oncological conditions. However, due to associated limitations like low oral bioavailability (14-39%), high inter/intra-subject variability, stability issues, etc., high doses (800 mg) are required, which further lead to non-specific toxicities and also contribute toward cancer resistance. Thus, to overcome these challenges, pazopanib-loaded PEGylated nanoliposomes were developed and evaluated against pancreatic cancer cell lines. The nanoliposomes were prepared by thin-film hydration method, followed by characterization and stability studies. This QbD-enabled process design successfully led to the development of a suitable pazopanib liposomal formulation with desirable properties. The % entrapment of PZP-loaded non-PEGylated and PEGylated nanoliposomes was found to be 75.2% and 84.9%, respectively, whereas their particle size was found to be 129.7 nm and 182.0 nm, respectively. The developed liposomal formulations exhibited a prolonged release and showed desirable physicochemical properties. Furthermore, these liposomal formulations were also assessed for in vitro cell lines, such as cell cytotoxicity assay and cell uptake. These studies confirm the effectiveness of developed liposomal formulations against pancreatic cancer cell lines. The outcomes of this work provide encouraging results and a way forward to thoroughly investigate its potential for PDAC treatment.
Collapse
Affiliation(s)
- Aishwarya Shinde
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Kanan Panchal
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Parameswar Patra
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Sonali Singh
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Sucharitha Enakolla
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India
| | - Rishi Paliwal
- Nanomedicine and Bioengineering Research Laboratory, Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | - Akash Chaurasiya
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Hyderabad, Pilani, India.
| |
Collapse
|
436
|
Nakamura H, Watanabe M, Takada K, Sato T, Hikage F, Umetsu A, Muramatsu J, Furuhashi M, Ohguro H. Modulation of Epithelial-Mesenchymal Transition Is a Possible Underlying Mechanism for Inducing Chemoresistance in MIA PaCa-2 Cells against Gemcitabine and Paclitaxel. Biomedicines 2024; 12:1011. [PMID: 38790973 PMCID: PMC11118094 DOI: 10.3390/biomedicines12051011] [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: 03/26/2024] [Revised: 04/23/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
To elucidate the currently unknown molecular mechanisms responsible for the similarity and difference during the acquirement of resistance against gemcitabine (GEM) and paclitaxel (PTX) in patients with pancreatic carcinoma, we examined two-dimensional (2D) and three-dimensional (3D) cultures of parent MIA PaCa-2 cells (MIA PaCa-2-PA) and their GEM resistance cell line (MIA PaCa-2-GR) and PTX resistance (MIA PaCa-2-PR). Using these cells, we examined 3D spheroid configurations and cellular metabolism, including mitochondrial and glycolytic functions, with a Seahorse bio-analyzer and RNA sequencing analysis. Compared to the MIA PaCa-2-PA, (1) the formation of the 3D spheroids of MIA PaCa-2-GR or -PR was much slower, and (2) their mitochondrial and glycolytic functions were greatly modulated in MIA PaCa-2-GR or -PR, and such metabolic changes were also different between their 2D and 3D culture conditions. RNA sequencing and bioinformatic analyses of the differentially expressed genes (DEGs) using an ingenuity pathway analysis (IPA) suggested that various modulatory factors related to epithelial -mesenchymal transition (EMT) including STAT3, GLI1, ZNF367, NKX3-2, ZIC2, IFIT2, HEY1 and FBLX, may be the possible upstream regulators and/or causal network master regulators responsible for the acquirement of drug resistance in MIA PaCa-2-GR and -PR. In addition, among the prominently altered DEGs (Log2 fold changes more than 6 or less than -6), FABP5, IQSEC3, and GASK1B were identified as unique genes associated with their antisense RNA or pseudogenes, and among these, FABP5 and GASK1B are known to function as modulators of cancerous EMT. Therefore, the observations reported herein suggest that modulations of cancerous EMT may be key molecular mechanisms that are responsible for inducing chemoresistance against GEM or PTX in MIA PaCa-2 cells.
Collapse
Affiliation(s)
- Hajime Nakamura
- Departments of Medical Oncology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (H.N.); (K.T.); (J.M.)
| | - Megumi Watanabe
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (A.U.)
| | - Kohichi Takada
- Departments of Medical Oncology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (H.N.); (K.T.); (J.M.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Fumihito Hikage
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (A.U.)
| | - Araya Umetsu
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (A.U.)
| | - Joji Muramatsu
- Departments of Medical Oncology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (H.N.); (K.T.); (J.M.)
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (M.F.)
| | - Hiroshi Ohguro
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (F.H.); (A.U.)
| |
Collapse
|
437
|
Moffat GT, Coyne Z, Albaba H, Aung KL, Dodd A, Espin-Garcia O, Moura S, Gallinger S, Kim J, Fraser A, Hutchinson S, Moulton CA, Wei A, McGilvray I, Dhani N, Jang R, Elimova E, Moore M, Prince R, Knox J. Impact of an Inter-Professional Clinic on Pancreatic Cancer Outcomes: A Retrospective Cohort Study. Curr Oncol 2024; 31:2589-2597. [PMID: 38785475 PMCID: PMC11119140 DOI: 10.3390/curroncol31050194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) presents significant challenges in diagnosis, staging, and appropriate treatment. Furthermore, patients with PDAC often experience complex symptomatology and psychosocial implications that require multi-disciplinary and inter-professional supportive care management from health professionals. Despite these hurdles, the implementation of inter-professional clinic approaches showed promise in enhancing clinical outcomes. To assess the effectiveness of such an approach, we examined the impact of the Wallace McCain Centre for Pancreatic Cancer (WMCPC), an inter-professional clinic for patients with PDAC at the Princess Margaret Cancer Centre (PM). Methods: This retrospective cohort study included all patients diagnosed with PDAC who were seen at the PM before (July 2012-June 2014) and after (July 2014-June 2016) the establishment of the WMCPC. Standard therapies such as surgery, chemotherapy, and radiation therapy remained consistent across both time periods. The cohorts were compared in terms of survival rates, disease stage, referral patterns, time to treatment, symptoms, and the proportion of patients assessed and supported by nursing and allied health professionals. Results: A total of 993 patients were included in the review, comprising 482 patients pre-WMCPC and 511 patients post-WMCPC. In the multivariate analysis, adjusting for ECOG (Eastern Cooperative Oncology Group) and stage, it was found that post-WMCPC patients experienced longer median overall survival (mOS, HR 0.84, 95% CI 0.72-0.98, p = 0.023). Furthermore, the time from referral to initial consultation date decreased significantly from 13.4 to 8.8 days in the post-WMCPC cohort (p < 0.001), along with a reduction in the time from the first clinic appointment to biopsy (14 vs. 8 days, p = 0.022). Additionally, patient-reported well-being scores showed improvement in the post-WMCPC cohort (p = 0.02), and these patients were more frequently attended to by nursing and allied health professionals (p < 0.001). Conclusions: The implementation of an inter-professional clinic for patients diagnosed with PDAC led to improvements in overall survival, patient-reported well-being, time to initial assessment visit and pathological diagnosis, and symptom management. These findings advocate for the adoption of an inter-professional clinic model in the treatment of patients with PDAC.
Collapse
Affiliation(s)
- Gordon Taylor Moffat
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Zachary Coyne
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Hamzeh Albaba
- Department of Oncology, Jack Ady Cancer Centre, University of Alberta, Lethbridge, AB T1J 1W5, Canada
| | - Kyaw Lwin Aung
- Livestrong Cancer Institutes and Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Anna Dodd
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Osvaldo Espin-Garcia
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1X6, Canada
| | - Shari Moura
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Steven Gallinger
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital Joseph, Toronto, ON M5G 1X5, Canada
- Toronto General Hospital, University Health Network, Toronto, ON M5G 2C4, Canada
| | - John Kim
- Department of Radiation Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Adriana Fraser
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Shawn Hutchinson
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Carol-Anne Moulton
- Toronto General Hospital, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Alice Wei
- Department of Surgery, Memorial Sloan Kettering Cancer Center, Weill-Cornell School of Medicine, Cornell University, New York City, NY 10065, USA
| | - Ian McGilvray
- Toronto General Hospital, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Neesha Dhani
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Raymond Jang
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Elena Elimova
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Malcolm Moore
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Rebecca Prince
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| | - Jennifer Knox
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 1X6, Canada
| |
Collapse
|
438
|
Ren J, Wu S, Su T, Ding J, Chen F, Li J, Wang Z, Han L, Wu Z. Analysis of chemoresistance characteristics and prognostic relevance of postoperative gemcitabine adjuvant chemotherapy in pancreatic cancer. Cancer Med 2024; 13:e7229. [PMID: 38698688 PMCID: PMC11066484 DOI: 10.1002/cam4.7229] [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: 11/09/2023] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
AIM To investigate the relationship between chemoresistance in pancreatic cancer patients receiving postoperative gemcitabine adjuvant therapy and specific clinical/pathological characteristics, as well as its impact on patient prognosis. METHODS From June 2018 to June 2021, clinical and pathological data of 148 pancreatic cancer patients were collected, and 101 patients were followed up for tumor recurrence/metastasis and survival status. The correlation between chemoresistance and specific clinical/pathological characteristics or patient prognosis was retrospectively analyzed. RESULTS Of the 148 patients, 78 were in the chemoresistance group and 70 in the non-chemoresistance group. Univariate analysis showed that the development of chemoresistance may be related to patient age, combined diabetes, preoperative CA19-9 level, tumor size, AJCC stage, vascular invasion, and positive lymph node ratio. Furthermore, subsequent multivariate analysis incorporating these variables indicated that tumor size may be a key factor influencing chemoresistance (p < 0.001, OR = 1.584). Log-rank test showed patients in the chemoresistance group had worse overall survival (OS) (HR = 2.102, p = 0.018) and progression free survival (PFS) (HR = 3.208, p = 0.002) than patients in the non-chemoresistance group; and patients with smaller size tumors (diameter ≤3 cm) had significantly better OS (HR = 2.923, p < 0.001) and PFS (HR = 2.930, p = 0.003) than those with larger size tumors (diameter >3 cm). CONCLUSIONS Patients with pancreatic cancer receiving postoperative gemcitabine adjuvant therapy are more likely to develop chemoresistance when their tumor sizes are larger (diameter >3 cm). Development of chemoresistance exacerbates the prognosis of patients with pancreatic cancer, and larger tumor size is also a risk factor for poor prognosis in these patients.
Collapse
Affiliation(s)
- Jiaqiang Ren
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shuai Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Tong Su
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Jiachun Ding
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fan Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jie Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Liang Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
439
|
Peng S, Huang H, Zhu X, Chen J, Ding X, Wang F, Chen L, Lu Z. Anlotinib plus tislelizumab for recurrent metastatic pancreas ductal adenocarcinoma with germline BRCA2 mutation: A case report. Exp Ther Med 2024; 27:178. [PMID: 38515651 PMCID: PMC10952340 DOI: 10.3892/etm.2024.12466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/09/2024] [Indexed: 03/23/2024] Open
Abstract
While combined immunotherapy and anti-angiogenic therapy have demonstrated efficacy in renal cell carcinoma, non-small cell lung cancer and hepatocellular carcinoma, the efficacy of first-line treatment for pancreatic ductal adenocarcinoma (PDAC) with germline BRCA2 mutation remains unproven. We described a BRCA2-mutated patient with PDAC who presented with posterior cardiac metastasis 8 months after surgery. After receiving four cycles of anlotinib combined with tislelizumab, abdominal CT scans indicated a complete response. The patient sustained this response for over 14 months on the combination regimen, with no reported adverse events. In conclusion, the combination of tislelizumab and anlotinib may offer a viable therapeutic option for recurrent metastatic BRCA2-mutated PDAC.
Collapse
Affiliation(s)
- Sujuan Peng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Hongxiang Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Xie Zhu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Jinhong Chen
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Xinjing Ding
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Fen Wang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Li Chen
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Zhihui Lu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
440
|
Magnan K, Williams L, Wang Q, Meade J. A Nine-Year-Old Child With Metastatic Pancreatic Adenocarcinoma. Cureus 2024; 16:e60670. [PMID: 38899258 PMCID: PMC11186403 DOI: 10.7759/cureus.60670] [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] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
Pancreatic ductal adenocarcinoma is exceedingly rare in children. Here, we report the case of a nine-year-old boy diagnosed with pancreatic ductal adenocarcinoma. The patient was treated per the National Comprehensive Cancer Network® (NCCN®) guidelines for adults with pancreatic cancer. Though the patient had multiple episodes of progression, the patient has remained alive with the disease 18 months after the initial diagnosis.
Collapse
Affiliation(s)
- Katelin Magnan
- Pediatrics, UPMC (University of Pittsburgh Medical Center) Children's Hospital of Pittsburgh, Pittsburgh, USA
| | - Linford Williams
- Medical Genetics, UPMC (University of Pittsburgh Medical Center) Children's Hospital of Pittsburgh, Pittsburgh, USA
| | - Qian Wang
- Department of Pathology, Children's Hospital of Pittsburgh, Pittsburgh, USA
| | - Julia Meade
- Pediatric Oncology, UPMC (University of Pittsburgh Medical Center), Pittsburgh, USA
| |
Collapse
|
441
|
Wehrli M, Guinn S, Birocchi F, Kuo A, Sun Y, Larson RC, Almazan AJ, Scarfò I, Bouffard AA, Bailey SR, Anekal PV, Llopis PM, Nieman LT, Song Y, Xu KH, Berger TR, Kann MC, Leick MB, Silva H, Salas-Benito D, Kienka T, Grauwet K, Armstrong TD, Zhang R, Zhu Q, Fu J, Schmidts A, Korell F, Jan M, Choi BD, Liss AS, Boland GM, Ting DT, Burkhart RA, Jenkins RW, Zheng L, Jaffee EM, Zimmerman JW, Maus MV. Mesothelin CAR T Cells Secreting Anti-FAP/Anti-CD3 Molecules Efficiently Target Pancreatic Adenocarcinoma and its Stroma. Clin Cancer Res 2024; 30:1859-1877. [PMID: 38393682 PMCID: PMC11062832 DOI: 10.1158/1078-0432.ccr-23-3841] [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: 12/15/2023] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
PURPOSE Targeting solid tumors with chimeric antigen receptor (CAR) T cells remains challenging due to heterogenous target antigen expression, antigen escape, and the immunosuppressive tumor microenvironment (TME). Pancreatic cancer is characterized by a thick stroma generated by cancer-associated fibroblasts (CAF), which may contribute to the limited efficacy of mesothelin-directed CAR T cells in early-phase clinical trials. To provide a more favorable TME for CAR T cells to target pancreatic ductal adenocarcinoma (PDAC), we generated T cells with an antimesothelin CAR and a secreted T-cell-engaging molecule (TEAM) that targets CAF through fibroblast activation protein (FAP) and engages T cells through CD3 (termed mesoFAP CAR-TEAM cells). EXPERIMENTAL DESIGN Using a suite of in vitro, in vivo, and ex vivo patient-derived models containing cancer cells and CAF, we examined the ability of mesoFAP CAR-TEAM cells to target PDAC cells and CAF within the TME. We developed and used patient-derived ex vivo models, including patient-derived organoids with patient-matched CAF and patient-derived organotypic tumor spheroids. RESULTS We demonstrated specific and significant binding of the TEAM to its respective antigens (CD3 and FAP) when released from mesothelin-targeting CAR T cells, leading to T-cell activation and cytotoxicity of the target cell. MesoFAP CAR-TEAM cells were superior in eliminating PDAC and CAF compared with T cells engineered to target either antigen alone in our ex vivo patient-derived models and in mouse models of PDAC with primary or metastatic liver tumors. CONCLUSIONS CAR-TEAM cells enable modification of tumor stroma, leading to increased elimination of PDAC tumors. This approach represents a promising treatment option for pancreatic cancer.
Collapse
Affiliation(s)
- Marc Wehrli
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Samantha Guinn
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Filippo Birocchi
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Adam Kuo
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Yi Sun
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Rebecca C. Larson
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Antonio J. Almazan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Irene Scarfò
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Amanda A. Bouffard
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Stefanie R. Bailey
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | | | | | - Linda T. Nieman
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Yuhui Song
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Katherine H. Xu
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Trisha R. Berger
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Michael C. Kann
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Mark B. Leick
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Blood and Marrow Transplant Program, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Harrison Silva
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Diego Salas-Benito
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Tamina Kienka
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Korneel Grauwet
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Todd D. Armstrong
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Rui Zhang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Qingfeng Zhu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Juan Fu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Andrea Schmidts
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Felix Korell
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Max Jan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - Bryan D. Choi
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - Andrew S. Liss
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Genevieve M. Boland
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - David T. Ting
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Richard A. Burkhart
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Russell W. Jenkins
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Elizabeth M. Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Jacquelyn W. Zimmerman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Marcela V. Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Blood and Marrow Transplant Program, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| |
Collapse
|
442
|
Wang S, Xiong Y, Luo Y, Shen Y, Zhang F, Lan H, Pang Y, Wang X, Li X, Zheng X, Lu X, Liu X, Cheng Y, Wu T, Dong Y, Lu Y, Cui J, Jia X, Yang S, Wang L, Wang Y. Genome-wide CRISPR screens identify PKMYT1 as a therapeutic target in pancreatic ductal adenocarcinoma. EMBO Mol Med 2024; 16:1115-1142. [PMID: 38570712 PMCID: PMC11099189 DOI: 10.1038/s44321-024-00060-y] [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: 10/25/2023] [Revised: 03/10/2024] [Accepted: 03/14/2024] [Indexed: 04/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with an overall 5-year survival rate of <12% due to the lack of effective treatments. Novel treatment strategies are urgently needed. Here, PKMYT1 is identified through genome-wide CRISPR screens as a non-mutant, genetic vulnerability of PDAC. Higher PKMYT1 expression levels indicate poor prognosis in PDAC patients. PKMYT1 ablation inhibits tumor growth and proliferation in vitro and in vivo by regulating cell cycle progression and inducing apoptosis. Moreover, pharmacological inhibition of PKMYT1 shows efficacy in multiple PDAC cell models and effectively induces tumor regression without overt toxicity in PDAC cell line-derived xenograft and in more clinically relevant patient-derived xenograft models. Mechanistically, in addition to its canonical function of phosphorylating CDK1, PKMYT1 functions as an oncogene to promote PDAC tumorigenesis by regulating PLK1 expression and phosphorylation. Finally, TP53 function and PRKDC activation are shown to modulate the sensitivity to PKMYT1 inhibition. These results define PKMYT1 dependency in PDAC and identify potential therapeutic strategies for clinical translation.
Collapse
Affiliation(s)
- Simin Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yangjie Xiong
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yuxiang Luo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yanying Shen
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Fengrui Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 200011, Shanghai, China
| | - Haoqi Lan
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yuzhi Pang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiaofang Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiaoqi Li
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Xufen Zheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiaojing Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiaoxiao Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yumei Cheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Tanwen Wu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yue Dong
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yuan Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 200011, Shanghai, China
| | - Jiujie Cui
- Department of Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Xiaona Jia
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China
| | - Sheng Yang
- Department of Oncology, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Liwei Wang
- Department of Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China.
| | - Yuexiang Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031, Shanghai, China.
| |
Collapse
|
443
|
Sagini MN, Zepp M, Eyol E, Ali DM, Gromova S, Dahlmann M, Behrens D, Groeschel C, Tischmeier L, Hoffmann J, Berger MR, Forssmann WG. EPI-X4, a CXCR4 antagonist inhibits tumor growth in pancreatic cancer and lymphoma models. Peptides 2024; 175:171111. [PMID: 38036098 DOI: 10.1016/j.peptides.2023.171111] [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: 08/06/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023]
Abstract
Endogenous peptide inhibitor for CXCR4 (EPI-X4) is a CXCR4 antagonist with potential for cancer therapy. It is a processed fragment of serum albumin from the hemofiltrate of dialysis patients. This study reports the efficacy of fifteen EPI-X4 derivatives in pancreatic cancer and lymphoma models. In vitro, the peptides were investigated for antiproliferation (cytotoxicity) by MTT assay. The mRNA expression for CXCR4 and CXCL12 was determined by RT-PCR, chip array and RNA sequencing. Chip array analysis yielded 634 genes associated with CXCR4/CXCL12 signaling. About 21% of these genes correlated with metastasis in the context of cell motility, proliferation, and survival. Expression levels of these genes were altered in pancreatic cancer (36%), lymphoma models (53%) and in patients' data (58%). EPI-X4 derivatives failed to inhibit cell proliferation due to low expression of CXCR4 in vitro, but inhibited tumor growth in the bioassays with significant efficacy. In the pancreatic cancer model, EPI-X4a, f and k inhibited mean tumor growth by > 50% and even caused complete remissions. In the lymphoma model, EPI-X4b, n and p inhibited mean tumor growth by > 70% and caused stable disease. Given the non-toxic and non-immunogenic properties of EPI-X4, these findings underscore its status as a promising therapy of pancreatic cancer and lymphoma and warrant further studies. SIMPLE SUMMARY: This study examined the value of chemokine receptor CXCR4 as an antineoplastic target for the endogenous peptide inhibitor of CXCR4 (EPI-X4), a 12-meric peptide derived from serum albumin. EPI-X4 inhibits CXCR4 interaction with its natural ligand, CXCL12 (SDF1). Therefore, malignancies (including pancreatic cancer and lymphoma) that depend on the CXCR4/CXCL12 pathway for progression can be targeted with EPI-X4. Of 634 genes that were linked to the CXCR4/CXCL12 pathway, 21% were associated with metastasis. In cultured human Suit2-007 pancreatic cancer cells, CXCR4 showed low to undetectable expression, which was why EPI-X4 did not inhibit pancreatic cancer cell proliferation. These findings were different in vivo, where CXCR4 was highly expressed and EPI-X4 inhibited tumor growth in rodents harboring pancreatic cancer or lymphoma. In the pancreatic cancer model, EPI-X4 derivatives a, f and k caused complete remissions, while in lymphomas EPI-X4 derivatives b, n and p caused stable disease.
Collapse
Affiliation(s)
- Micah N Sagini
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Michael Zepp
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Ergül Eyol
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Doaa M Ali
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Svetlana Gromova
- EPO, Experimental Pharmacology & Oncology Berlin-Buch GmbH, Germany
| | - Mathias Dahlmann
- EPO, Experimental Pharmacology & Oncology Berlin-Buch GmbH, Germany
| | - Diana Behrens
- EPO, Experimental Pharmacology & Oncology Berlin-Buch GmbH, Germany
| | - Christian Groeschel
- NeoPep Pharma GmbH & Co. KG., Hannover, Germany and Hannover Medical School, Department of Internal Medicine, Germany
| | - Linus Tischmeier
- NeoPep Pharma GmbH & Co. KG., Hannover, Germany and Hannover Medical School, Department of Internal Medicine, Germany
| | - Jens Hoffmann
- EPO, Experimental Pharmacology & Oncology Berlin-Buch GmbH, Germany
| | - Martin R Berger
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Heidelberg, Germany.
| | - Wolf-Georg Forssmann
- NeoPep Pharma GmbH & Co. KG., Hannover, Germany and Hannover Medical School, Department of Internal Medicine, Germany.
| |
Collapse
|
444
|
Robbins KJ, Newcomer KF, Barnell EK, Anzelmo MA, Liu J, Hawkins WG. Neoadjuvant Chemotherapy is Associated with Increased Risk of Postoperative DVT After Distal Pancreatectomy for Pancreatic Adenocarcinoma: a NSQIP Analysis. Ann Surg Oncol 2024; 31:2873-2881. [PMID: 38151621 DOI: 10.1245/s10434-023-14763-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 11/25/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Venous thromboembolism (VTE) remains a persistent source of postoperative morbidity despite prevention and mitigation efforts. Cancer, surgery, and chemotherapy are known risk factors for VTE. Existing literature suggests that neoadjuvant therapy (NAT) may contribute to increased VTE risk in the postoperative period, but few authors specifically examine this relationship in distal pancreatic adenocarcinoma (PDAC). In this study, we analyze the association of NAT and postoperative VTE in patients who underwent distal pancreatectomy (DP) for PDAC. PATIENTS AND METHODS Using the American College of Surgeons (ACS) National Surgical Quality Improvement Program (NSQIP) database, we analyzed the Procedure Targeted files for pancreatectomy from 2014 to 2020. Adults with PDAC who underwent DP were grouped by receipt of NAT. The primary outcome was the rate of deep venous thrombosis (DVT) and the secondary outcome was the rate of pulmonary embolism (PE). We performed univariate and multivariate logistic regression analysis to determine risk factors associated with postoperative DVT. RESULTS There were 4327 patients with PDAC who underwent DP. Of these, 1414 (32.7%) had NAT. Receipt of NAT was significantly associated with postoperative DVT requiring therapy (3.5% vs. 2.3%, p = 0.02), but was not associated with PE (p = 0.42). On MVA, NAT was associated with a 73% greater chance of developing postoperative DVT [odds ratio (OR) 1.73, 95% CI 1.18-2.55]. CONCLUSIONS Patients who receive NAT prior to DP for PDAC are 73% more likely to develop postoperative DVT compared with upfront resection. As NAT becomes more commonplace, these high-risk patients should be prioritized for guideline-recommended extended duration prophylaxis.
Collapse
Affiliation(s)
- Keenan J Robbins
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Kenneth F Newcomer
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Erica K Barnell
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Jingxia Liu
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - William G Hawkins
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
- Alvin J. Siteman Cancer Center, St. Louis, MO, USA.
| |
Collapse
|
445
|
Tentes AA, Kyziridis D, Kalakonas A, Iliadis A, Fotiadou A. Pancreatic cancer with synchronous peritoneal and hepatic metastases: A case report. Int J Surg Case Rep 2024; 118:109588. [PMID: 38581939 PMCID: PMC11015438 DOI: 10.1016/j.ijscr.2024.109588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 04/08/2024] Open
Abstract
Introduction and importance: There is evidence that patients with limited peritoneal carcinomatosis of pancreatic cancer or those with low burden of hepatic metastases are amenable to surgical resection. A case report of a patient with cancer of the pancreatic tail and synchronous peritoneal and hepatic metastases is presented. CASE PRESENTATION A male patient, 66 years old, underwent cytoreductive surgery (CRS) plus hyperthermic intraperitoneal chemotherapy (HIPEC) and radio-frequency ablation (RFA) for synchronous hepatic metastases simultaneously to distal pancreatectomy for adenocarcinoma of the pancreas. Adjuvant chemotherapy followed the R0 surgery. The patient remained disease free for 18 months, developed liver recurrence and died 28 months after the initial operation. DISCUSSION CRS plus HIPEC with synchronous ablation or resection of hepatic metastases may be used for the treatment of pancreatic cancer with synchronous peritoneal and hepatic metastases in highly selected patients. CONCLUSION Further studies are needed to confirm whether patients with synchronous peritoneal and hepatic metastases are offered survival benefit from complex surgical intervention (CRS plus HIPEC combined with hepatic resection or RFA).
Collapse
Affiliation(s)
| | | | | | - Alexandros Iliadis
- Laboratory of Diagnostic Histopathology, Mitropoleos 115, Thessaloniki 54622, Greece
| | - Anastasia Fotiadou
- Laboratory of Diagnostic Histopathology, Mitropoleos 115, Thessaloniki 54622, Greece
| |
Collapse
|
446
|
Mendis S, Lipton L, To YH, Ananda S, Michael M, McLachlan SA, Thomson B, Loveday B, Knowles B, Fox A, Nikfarjam M, Usatoff V, Shapiro J, Clarke K, Pattison S, Chee CE, Zielinski R, Wong R, Gibbs P, Lee B. Early onset pancreatic cancer-exploring contemporary treatment and outcomes using real-world data. Br J Cancer 2024; 130:1477-1484. [PMID: 38448752 PMCID: PMC11058801 DOI: 10.1038/s41416-024-02619-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Pancreatic cancer incidence is increasing in younger populations. Differences between early onset pancreatic cancer (EOPC) and later onset pancreatic cancer (LOPC), and how these should inform management warrant exploration in the contemporary setting. METHODS A prospectively collected multi-site dataset on consecutive pancreatic adenocarcinoma patients was interrogated. Patient, tumour, treatment, and outcome data were extracted for EOPC (≤50 years old) vs LOPC (>50 years old). RESULTS Of 1683 patients diagnosed between 2016 and 2022, 112 (6.7%) were EOPC. EOPC more frequently had the tail of pancreas tumours, earlier stage disease, surgical resection, and trended towards increased receipt of chemotherapy in the curative setting compared to LOPC. EOPC more frequently received 1st line chemotherapy, 2nd line chemotherapy, and chemoradiotherapy than LOPC in the palliative setting. Recurrence-free survival was improved for the tail of pancreas EOPC vs LOPC in the resected setting; overall survival was superior for EOPC compared to LOPC across the resected, locally advanced unresectable and metastatic settings. CONCLUSIONS EOPC remains a small proportion of pancreatic cancer diagnoses. The more favourable outcomes in EOPC suggest these younger patients are overall deriving benefits from increased treatment in the curative setting and increased therapy in the palliative setting.
Collapse
Affiliation(s)
- Shehara Mendis
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
| | | | - Yat Hang To
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Sumitra Ananda
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Michael Michael
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Sue-Anne McLachlan
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, St Vincent's Hospital, Fitzroy, VIC, Australia
| | - Benjamin Thomson
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Benjamin Loveday
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Brett Knowles
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Epworth Healthcare, Melbourne, VIC, Australia
| | - Adrian Fox
- Department of Hepatobiliary Surgery, St Vincent's Hospital, Fitzroy, VIC, Australia
| | - Mehrdad Nikfarjam
- University of Melbourne, Parkville, VIC, Australia
- Department of Hepatobiliary Surgery, Austin Health, Heidelberg, VIC, Australia
| | | | - Julia Shapiro
- Department of Medicine, Alfred Hospital, Prahran, VIC, Australia
| | - Kate Clarke
- Department of Medical Oncology, Wellington Hospital, Wellington, New Zealand
| | - Sharon Pattison
- Department of Medicine, Dunedin School of Medicine, University of Otago, Otago, New Zealand
| | - Cheng Ean Chee
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Rob Zielinski
- Department of Medical Oncology, Orange Hospital, Orange, NSW, Australia
- Department of Medical Oncology, Dubbo Base Hospital, Dubbo, NSW, Australia
- Department of Medical Oncology, Bathurst Base Hospital, West Bathurst, NSW, Australia
| | - Rachel Wong
- Epworth Healthcare, Melbourne, VIC, Australia
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
- Department of Medical Oncology, Eastern Health, Box Hill, VIC, Australia
| | - Peter Gibbs
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
| | - Belinda Lee
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Department of Medical Oncology, Northern Hospital, Epping, VIC, Australia
| |
Collapse
|
447
|
Girolimetti G, Pelisenco IA, Eusebi LH, Ricci C, Cavina B, Kurelac I, Verri T, Calcagnile M, Alifano P, Salvi A, Bucci C, Guerra F. Dysregulation of a Subset of Circulating and Vesicle-Associated miRNA in Pancreatic Cancer. Noncoding RNA 2024; 10:29. [PMID: 38804361 PMCID: PMC11130804 DOI: 10.3390/ncrna10030029] [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: 02/20/2024] [Revised: 04/14/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive neoplasia, characterized by early metastasis, low diagnostic rates at early stages, resistance to drugs, and poor prognosis. There is an urgent need to better characterize this disease in order to identify efficient diagnostic/prognostic biomarkers. Since microRNAs (miRNAs) contribute to oncogenesis and metastasis formation in PDAC, they are considered potential candidates for fulfilling this task. In this work, the levels of two miRNA subsets (involved in chemoresistance or with oncogenic/tumor suppressing functions) were investigated in a panel of PDAC cell lines and liquid biopsies of a small cohort of patients. We used RT-qPCR and droplet digital PCR (ddPCR) to measure the amounts of cellular- and vesicle-associated, and circulating miRNAs. We found that both PDAC cell lines, also after gemcitabine treatment, and patients showed low amounts of cellular-and vesicle-associated miR-155-5p, compared to controls. Interestingly, we did not find any differences when we analyzed circulating miR-155-5p. Furthermore, vesicle-related miR-27a-3p increased in cancer patients compared to the controls, while circulating let-7a-5p, miR-221-3p, miR-23b-3p and miR-193a-3p presented as dysregulated in patients compared to healthy individuals. Our results highlight the potential clinical significance of these analyzed miRNAs as non-invasive diagnostic molecular tools to characterize PDAC.
Collapse
Affiliation(s)
- Giulia Girolimetti
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (G.G.); (T.V.); (M.C.); (F.G.)
| | - Iulia Andreea Pelisenco
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (I.A.P.); (A.S.)
| | - Leonardo Henry Eusebi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (L.H.E.); (C.R.); (B.C.); (I.K.)
- Gastroenterology Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Claudio Ricci
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (L.H.E.); (C.R.); (B.C.); (I.K.)
- Pancreatic Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Beatrice Cavina
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (L.H.E.); (C.R.); (B.C.); (I.K.)
- Centre for Applied Biomedical Research (CRBA), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Ivana Kurelac
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (L.H.E.); (C.R.); (B.C.); (I.K.)
- Centre for Applied Biomedical Research (CRBA), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Tiziano Verri
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (G.G.); (T.V.); (M.C.); (F.G.)
| | - Matteo Calcagnile
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (G.G.); (T.V.); (M.C.); (F.G.)
| | - Pietro Alifano
- Department of Experimental Medicine (DiMeS), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy;
| | - Alessandro Salvi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (I.A.P.); (A.S.)
| | - Cecilia Bucci
- Department of Experimental Medicine (DiMeS), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy;
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (G.G.); (T.V.); (M.C.); (F.G.)
| |
Collapse
|
448
|
Weiss L, Heinemann V, Fischer LE, Gieseler F, Hoehler T, Mayerle J, Quietzsch D, Reinacher-Schick A, Schenk M, Seipelt G, Siveke JT, Stahl M, Vehling-Kaiser U, Waldschmidt DT, Dorman K, Zhang D, Westphalen CB, von Bergwelt-Baildon M, Boeck S, Haas M. Three-month life expectancy as inclusion criterion for clinical trials in advanced pancreatic cancer: is it really a valid tool for patient selection? Clin Transl Oncol 2024; 26:1268-1272. [PMID: 37794220 PMCID: PMC11026194 DOI: 10.1007/s12094-023-03323-1] [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: 07/05/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023]
Abstract
PURPOSE To analyze the 3-month life expectancy rate in pancreatic cancer (PC) patients treated within prospective trials from the German AIO study group. PATIENTS AND METHODS A pooled analysis was conducted for patients with advanced PC that were treated within five phase II/III studies conducted between 1997 and 2017 (Gem/Cis, Ro96, RC57, ACCEPT, RASH). The primary goal for the current report was to identify the actual 3-month survival rate, a standard inclusion criterion in oncology trials. RESULTS Overall, 912 patients were included, 83% had metastatic and 17% locally advanced PC; the estimated median overall survival (OS) was 7.1 months. Twenty-one percent of the participants survived < 3 months, with a range from 26% in RC57 to 15% in RASH. Significant predictors for not reaching 3-month OS were > 1 previous treatment line (p < 0.001) and performance status (p < 0.001). CONCLUSIONS Despite the definition of a life expectancy of > 3 months as a standard inclusion criterion in clinical trials for advanced PC, a significant proportion of study patients does not survive > 3 months. TRIAL REGISTRATION NUMBERS NCT00440167 (AIO-PK0104), NCT01729481 (RASH), NCT01728818 (ACCEPT).
Collapse
Affiliation(s)
- Lena Weiss
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany
| | - Volker Heinemann
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Laura E Fischer
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany
| | - Frank Gieseler
- Department of Hematology and Oncology, University Hospital Schleswig-Holstein-Campus Lübeck, Lübeck, Germany
| | - Thomas Hoehler
- Department of Medicine I, Prosper Hospital, Recklinghausen, Germany
| | - Julia Mayerle
- Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Detlef Quietzsch
- Department of Medical Oncology, Klinikum Chemnitz, Chemnitz, Germany
| | - Anke Reinacher-Schick
- Department of Hematology and Oncology, St. Josef Hospital, Ruhr University, Bochum, Germany
| | - Michael Schenk
- Department of Hematology and Oncology, Klinikum Barmherzige Brüder, Regensburg, Germany
| | | | - Jens T Siveke
- West German Cancer Center, Bridge Institute of Experimental Tumor Therapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Michael Stahl
- Department of Medical Oncology, Evang. Kliniken Essen-Mitte, Essen, Germany
| | | | - Dirk T Waldschmidt
- Department of Gastroenterology and Hepatology, University of Cologne, Cologne, Germany
| | - Klara Dorman
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Danmei Zhang
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - C Benedikt Westphalen
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Stefan Boeck
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Michael Haas
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany.
- Comprehensive Cancer Center, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
449
|
Yoon H, Shin Y, Ryoo BY, Jeong H, Park I, Seo DW, Lee SS, Park DH, Song TJ, Oh D, Hwang DW, Lee JH, Song KB, Park Y, Kwak BJ, Hong SM, Park JH, Kim SC, Kim KP, Yoo C. Clinical outcomes of second-line therapy following disease progression on first-line modified FOLFIRINOX for borderline resectable and locally advanced pancreatic adenocarcinoma. Pancreatology 2024; 24:424-430. [PMID: 38395676 DOI: 10.1016/j.pan.2024.02.004] [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: 11/18/2023] [Revised: 01/21/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Modified FOLFIRINOX (mFOLFIRINOX) is one of the standard first-line therapies in borderline resectable pancreatic cancer (BRPC) and locally advanced unresectable pancreatic cancer (LAPC). However, there is no globally accepted second-line therapy following progression on mFOLFIRINOX. METHODS Patients with BRPC and LAPC (n = 647) treated with first-line mFOLFIRINOX between January 2017 and December 2020 were included in this retrospective analysis. The details of the treatment outcomes and patterns of subsequent therapy after mFOLFIRINOX were reviewed. RESULTS With a median follow-up duration of 44.2 months (95% confidence interval [CI], 42.3-47.6), 322 patients exhibited disease progression on mFOLFIRINOX-locoregional progression only in 177 patients (55.0%) and distant metastasis in 145 patients (45.0%). The locoregional progression group demonstrated significantly longer post-progression survival (PPS) than that of the distant metastasis group (10.1 vs. 7.3 months, p = 0.002). In the locoregional progression group, survival outcomes did not differ between second-line chemoradiation/radiotherapy and systemic chemotherapy (progression-free survival with second-line therapy [PFS-2], 3.2 vs. 4.3 months; p = 0.649; PPS, 10.7 vs. 10.2 months; p = 0.791). In patients who received second-line systemic chemotherapy following progression on mFOLFIRINOX (n = 211), gemcitabine plus nab-paclitaxel was associated with better disease control rates (69.2% vs. 42.3%, p = 0.005) and PFS-2 (3.8 vs. 1.7 months, p = 0.035) than gemcitabine monotherapy. CONCLUSIONS The current study showed the real-world practice pattern of subsequent therapy and clinical outcomes following progression on first-line mFOLFIRINOX in BRPC and LAPC. Further investigation is necessary to establish the optimal therapy after failure of mFOLFIRINOX.
Collapse
Affiliation(s)
- Hyunseok Yoon
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yeokyeong Shin
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Baek-Yeol Ryoo
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyehyun Jeong
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Inkeun Park
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dong-Wan Seo
- Departments of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sang Soo Lee
- Departments of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Do Hyun Park
- Departments of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Tae Jun Song
- Departments of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dongwook Oh
- Departments of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dae Wook Hwang
- Departments of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae Hoon Lee
- Departments of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ki Byung Song
- Departments of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yejong Park
- Departments of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Bong Jun Kwak
- Departments of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seung-Mo Hong
- Departments of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jin-Hong Park
- Departments of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Song Cheol Kim
- Departments of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Kyu-Pyo Kim
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Changhoon Yoo
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
450
|
Frimpong E, Bulusu R, Okoro J, Inkoom A, Ndemazie N, Rogers S, Zhu X, Han B, Agyare E. Development of novel pyrimidine nucleoside analogs as potential anticancer agents: Synthesis, characterization, and In-vitro evaluation against pancreatic cancer. Eur J Pharm Sci 2024; 196:106754. [PMID: 38554983 PMCID: PMC11229414 DOI: 10.1016/j.ejps.2024.106754] [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: 01/24/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
The present study proposed modification of 5-FU by conjugation with an acyl chloride and a 5-membered heterocyclic ring to improve its in-vitro cytotoxicity and metabolic stability. XYZ-I-71 and XYZ-I-73 were synthesized by introducing a tetrahydrofuran ring on 5-fluorocytosine (a precursor of 5-FU) and conjugation with octanoyl chloride and lauroyl chloride, respectively. The structure of the synthesized compounds was validated using NMR and micro-elemental analysis. The antiproliferative activity of the analogs was determined against MiaPaCa-2, PANC-1, and BxPC-3 pancreatic cancer cells. The analog's stability in human liver microsomes was quantified by HPLC. We found that the XYZ-I-73 (IC50 3.6 ± 0.4 μM) analog was most effective against MiaPaCa-2 cells compared to XYZ-I-71(IC50 12.3 ± 1.7 μM), GemHCl (IC50 24.2 ± 1.3 μM), Irinotecan (IC50 10.1 ± 1.5 μM) and 5-FU (IC50 13.2 ± 1.1 μM). The antiproliferative effects of this analog in Miapaca-2 cells is evident based on it having a 7-fold,3-fold, and 4-fold increased cytotoxic effect over Gem-HCl, Irinotecan, and 5-FU, respectively. On the other hand, XYZ-I-71 exhibited a 2-fold increased cytotoxic effect over Gem-HCl but a comparable cytotoxic effect to 5-FU and Irinotecan in MiaPaCa-2 cells. A similar trend of higher XYZ-I-73 inhibition was observed in PANC-1 and BxPC-3 cultures. For 48-h MiaPaCa-2 cell migration studies, XYZ-I-73 (5 μM) significantly reduced migration (# of migrated cells, 168 ± 2.9), followed by XYZ-I-71(315±2.1), Gem-HCl (762±3.1) and 5-FU (710 ± 3.2). PARP absorbance studies demonstrated significant inhibition of PARP expression of XYZ-I-73 treated cells compared to 5-FU, GemHCl, and XYZ-I-71. Further, BAX and p53 expressions were significantly increased in cells treated with XYZ-I-73 compared to 5-FU, GemHCl, and XYZ-I-71. In-vitro, metabolic stability studies showed that 80 ± 5.9% of XYZ-I-71 and XYZ-I-73 remained intact after 2 h exposure in liver microsomal solution compared to 5-FU. The XYZ-I-73 analog demonstrated a remarkable cytotoxic effect and improved in-vitro metabolic stability over the selected standard drugs and may have potential anticancer activity against pancreatic cancer.
Collapse
Affiliation(s)
- Esther Frimpong
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Raviteja Bulusu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Joy Okoro
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Andriana Inkoom
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Nkafu Ndemazie
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States; Department of Internal Medicine, Richmond University Medical Center, Staten Island, NY, United States
| | - Sherise Rogers
- Department of Medicine, Division of Hematology and Oncology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Xue Zhu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Bo Han
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States.
| |
Collapse
|