151
|
Lai X, Cheng D, Xu H, Wang J, Lv X, Yao H, Li L, Wu J, Ye S, Li Z. Phase I Trial of Upamostat Combined With Gemcitabine in Locally Unresectable or Metastatic Pancreatic Cancer: Safety and Preliminary Efficacy Assessment. Cancer Med 2025; 14:e70550. [PMID: 39739976 DOI: 10.1002/cam4.70550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
AIM This study aimed to determine the maximum tolerated dose (MTD) of the urokinase plasminogen activator (uPA) inhibitor upamostat (LH011) in combination with gemcitabine for locally advanced unresectable or metastatic pancreatic cancer. METHOD Seventeen patients were enrolled and received escalating doses of oral LH011 (100, 200, 400, or 600 mg) daily alongside 1000 mg/m2 of gemcitabine. Safety profiles, tumor response (including response rate and progression-free survival), pharmacokinetics, and changes in CA199 and D-dimer levels were assessed. RESULTS During the study period (Day0-Day49), no patients achieved partial response. Stable disease (SD) was observed in 12 patients (70.6%), while four patients (23.5%) experienced progressive disease (PD). One patient withdrew due to a serious adverse event (SAE) on D47. Pharmacokinetic analysis revealed a dose-related increase in LH011 and its metabolite WX-UK1 exposure from 100 to 400 mg but not in the 600 mg group. Hematological toxicity, mainly attributable to gemcitabine, was the predominant grade 3 or 4 adverse event, with additional occurrences of loss of appetite, rash, and interstitial lung disease. Sinus bradycardia possibly linked to LH011 rather than gemcitabine was noted. The MTD was not reached. CONCLUSION Combining LH011 at doses ranging from 100 to 600 mg with gemcitabine every 21 days demonstrated manageable safety and tolerability. However, tumor response did not significantly differ among the dose groups, suggesting the need for further investigation. TRIAL REGISTRATION NCT05329597.
Collapse
Affiliation(s)
- Xiuping Lai
- Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Di Cheng
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huixin Xu
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingshu Wang
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaozhi Lv
- Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Herui Yao
- Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liuning Li
- Department of Medical Oncology, GuangDong Province Hospital of Chinese Medicine, the Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, China
| | - Junyan Wu
- Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Suiwen Ye
- Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihua Li
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
152
|
Reni M, Giordano G, Audisio M, Orsi G, Macchini M, Gobba SM, Rapposelli I, Lucenti A, Luchena G, Faloppi L, Zustovich F, Ricci V, Cergnul M, Formica V, Procaccio L, Baccolini V, Briccolani A, Cascinu S, Peretti U. Exploring external validity of chemotherapy for pancreatic ductal adenocarcinoma in real life. Dig Liver Dis 2025; 57:104-110. [PMID: 39003164 DOI: 10.1016/j.dld.2024.06.025] [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: 03/21/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/15/2024]
Abstract
INTRODUCTION Cisplatin, nab-paclitaxel, capecitabine, and gemcitabine (PAXG) regimen activity was assessed in a single institution phase II trial (PACT-19) on pancreatic ductal adenocarcinoma (PDAC). The PACT-31 study explored the external validity of PACT-19 results. MATERIALS AND METHODS Patients aged ≥18 and ≤75 years with KPS ≥70, and PDAC diagnosis receiving PAXG in the participating institutions were eligible and categorized as follows: A) PACT-19; B) PACT-31-HSR; C) PACT-31-non-HSR. With a sample of 175 patients, assuming a target 1-year overall survival of 60 % for metastatic and of 80 % for non-metastatic patients, the trial will be considered successful with the 1-year OS falling into the 95 % CI. RESULTS Data from 68 PACT-19 and 168 PACT-31 patients were retrieved. After 124 events, 1yOS was 52.5 % (95 %CI: 44.6-60.4 %) for metastatic and 80.5 % (95 %CI: 71.9-89.1 %) for non-metastatic patients. Survival overlapped between PACT-19 and PACT-31-HSR (median 17.6 and 17.4 months, p = 0.21) and was significantly shorter in PACT-31-non-HSR (median 11.3 months; p = 0.03). Differences of dose-intensity, use of maintenance therapy, and treatment after progression between PACT-31-HSR and non-HSR were evidenced. DISCUSSION PACT-19 results have external validity. The outcome difference between HSR and non-HSR centers endorses the need of creating a hub-and-spoke network aimed at sharing the expertise on rare-diseases.
Collapse
Affiliation(s)
- Michele Reni
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy.
| | - Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy, Policlinico Ospedali Riuniti di Foggia, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 251, 71122 Foggia, Italy
| | - Marco Audisio
- Department of Oncology, University of Turin, AO Ordine Mauriziano, Medical Oncology Unit, ASL Torino 4, Ivrea, Turin, Italy
| | - Giulia Orsi
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| | - Marina Macchini
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| | - Stefania Maria Gobba
- Department of Medical Oncology, ASST-Settelaghi, Ospedale di Circolo, Viale Luigi Borri, 57, 21100 Varese, Italy
| | - Ilario Rapposelli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via Piero Maroncelli, 40, 47014 Meldola, (FC), Italy
| | - Antonio Lucenti
- U.O.C. Oncologia Medica Ospedale Maria Paternò Arezzo, 97100, Maria Paternò E Arezzo, Ragusa, Italy
| | - Giovanna Luchena
- U.O.C. Oncologia, Ospedale S. Anna ASST-LARIANA, Via Ravona, 1, 22020 San Fermo della Battaglia, Como, Italy
| | - Luca Faloppi
- Medical Oncology Unit, Azienda Sanitaria Territoriale di Macerata, Via Santa Lucia, 2, Macerata, Italy
| | - Fable Zustovich
- Department of Medical Oncology, AULSS 1 Dolomiti, Ospedale S.Martino, V.le Europa, 22, 32100 Belluno, Italy
| | - Vincenzo Ricci
- Medical Oncology Unit, Azienda Ospedaliera di Rilievo Nazionale 'San Pio', Via Dell'Angelo 1, Benevento, Italy
| | - Massimiliano Cergnul
- Ospedale Civile di Legnano, ASST OVEST MILANESE, Via Candiani, 2, 20025 Legnano, Italy
| | - Vincenzo Formica
- Medical Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Viale Oxford, 81, 00133 Roma, Italy
| | - Letizia Procaccio
- Dept of Oncology Veneto Institute of Oncology IOV, IRCCS, Via Gattamelata, 64, 35128 Padova, Italy
| | - Valeria Baccolini
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| | - Assunta Briccolani
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| | - Stefano Cascinu
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| | - Umberto Peretti
- Department of Medical Oncology IRCCS Ospedale San Raffaele, Vita e Salute University, Via Olgettina 60, 20132 Milan, Italy
| |
Collapse
|
153
|
Luan X, Wang X, Bian G, Li X, Gao Z, Liu Z, Zhang Z, Han T, Zhao J, Zhao H, Luan X, Zhu W, Dong L, Guo F. Exosome applications for the diagnosis and treatment of pancreatic ductal adenocarcinoma: An update (Review). Oncol Rep 2025; 53:13. [PMID: 39575479 PMCID: PMC11605277 DOI: 10.3892/or.2024.8846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a malignant neoplasm that typically manifests with subtle clinical manifestations in its early stages and frequently eludes diagnosis until the advanced phases of the disease. The limited therapeutic options available for PDAC significantly contribute to its high mortality rate, highlighting the urgent need for novel biomarkers capable of effectively identifying early clinical manifestations and facilitating precise diagnosis. The pivotal role of cellular exosomes in both the pathogenesis and therapeutic interventions for PDAC has been underscored. Furthermore, researchers have acknowledged the potential of exosomes as targeted drug carriers against regulatory cells in treating PDAC. The present article aims to provide a comprehensive review encompassing recent advancements in utilizing exosomes for elucidating mechanisms underlying disease development, patterns of metastasis, diagnostic techniques and treatment strategies associated with PDAC.
Collapse
Affiliation(s)
- Xinchi Luan
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xuezhe Wang
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Gang Bian
- Department of Gastroenterology, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Xiaoxuan Li
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266031, P.R. China
| | - Ziru Gao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Zijiao Liu
- School of Clinical and Basic Medicine and Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Zhishang Zhang
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Tianyue Han
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Jinpeng Zhao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Hongjiao Zhao
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xinyue Luan
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Wuhui Zhu
- Department of Hepatobiliary surgery, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Lili Dong
- Department of Gastroenterology, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong 266041, P.R. China
| | - Feifei Guo
- Pathophysiology Department, School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
154
|
Hashimoto D, Satoi S, Yamaki S, Nakayama S, Shibata N, Matsumura K, Miyazaki H, Matsui Y, Tsybulskyi D, Sang NT, Ikeura T, Kanai M, Sekimoto M. Neoadjuvant Treatment with Changes in Chemotherapy Regimens According to Carbohydrate Antigen 19-9 Level for Resectable/Borderline Resectable Pancreatic Ductal Adenocarcinoma. Ann Surg Oncol 2025; 32:517-528. [PMID: 39433718 DOI: 10.1245/s10434-024-16361-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: 03/28/2024] [Accepted: 09/28/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND The response of carbohydrate antigen (CA) 19-9 to neoadjuvant therapy (NAT) for pancreatic ductal adenocarcinoma (PDAC) may contribute to outcomes. This study aimed to investigate the effect of changes in NAT regimens based on CA19-9 level. METHODS This single-center retrospective study included patients with resectable/borderline resectable (R/BR)-PDAC undergoing NAT from 2008 to 2022. A CA19-9 level lower than 150 IU/mL after NAT was the criterion for resection. If the level did not decrease, the chemotherapy regimen was changed to satisfy the criterion. The patient cohort was divided into group A (satisfied criterion without changing chemotherapy), group B (did not receive chemotherapy change, could not satisfy the criterion), group C (received chemotherapy change, satisfied the criterion), and group D (received chemotherapy change, could not satisfy the criterion). RESULTS The study cohort included 283 patients. After first-line chemotherapy, 112 (39.6%) patients did not satisfy the criterion (groups B [n = 64], C [n = 32], and D [n = 16]). Of the 283 patients, 48 (17%) received a chemotherapy change (groups C and D). The patients in groups C and D showed significantly better overall survival (OS, 35.9 months) than the group B patients (25.7 months) (P = 0.035). The OS of the group C patients (63.8 months) was similar to the OS of the group A patients (n = 171: 56.3 months; P = 0.430). Multivariate analysis of the patients in groups B, C, and D identified chemotherapy change as an independent prognostic factor for OS and progression-free survival. CONCLUSION Changing the chemotherapy targeting the CA19-9 level can improve the outcome of R/BR-PDAC patients with poor biologic response to first-line NAT.
Collapse
Affiliation(s)
- Daisuke Hashimoto
- Department of Surgery, Kansai Medical University, Hirakata City, Osaka, Japan
| | - Sohei Satoi
- Department of Surgery, Kansai Medical University, Hirakata City, Osaka, Japan.
- Division of Surgical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - So Yamaki
- Department of Surgery, Kansai Medical University, Hirakata City, Osaka, Japan
| | - Shinji Nakayama
- Third Department of Internal Medicine, Kansai Medical University, Hirakata City, Osaka, Japan
| | - Nobuhiro Shibata
- Cancer Treatment Center, Kansai Medical University, Hirakata City, Osaka, Japan
| | - Kazuki Matsumura
- Department of Surgery, Kansai Medical University, Hirakata City, Osaka, Japan
| | - Hidetaka Miyazaki
- Department of Surgery, Kansai Medical University, Hirakata City, Osaka, Japan
| | - Yuki Matsui
- Department of Surgery, Kansai Medical University, Hirakata City, Osaka, Japan
| | - Denys Tsybulskyi
- Department of Surgery, Kansai Medical University, Hirakata City, Osaka, Japan
| | - Nguyen Thanh Sang
- Department of Surgery, Kansai Medical University, Hirakata City, Osaka, Japan
| | - Tsukasa Ikeura
- Third Department of Internal Medicine, Kansai Medical University, Hirakata City, Osaka, Japan
| | - Masashi Kanai
- Cancer Treatment Center, Kansai Medical University, Hirakata City, Osaka, Japan
| | - Mitsugu Sekimoto
- Department of Surgery, Kansai Medical University, Hirakata City, Osaka, Japan
| |
Collapse
|
155
|
Nam H, Lee W, Lee YJ, Kim JM, Jung KH, Hong SS, Kim SC, Park S. Taurine Synthesis by 2-Aminoethanethiol Dioxygenase as a Vulnerable Metabolic Alteration in Pancreatic Cancer. Biomol Ther (Seoul) 2025; 33:143-154. [PMID: 39637922 PMCID: PMC11704412 DOI: 10.4062/biomolther.2024.086] [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: 05/27/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 12/07/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) exhibits an altered metabolic profile compared to normal pancreatic tissue. However, studies on actual pancreatic tissues are limited. Untargeted metabolomics analysis was conducted on 54 pairs of tumor and matched normal tissues. Taurine levels were validated via immunohistochemistry (IHC) on separate PDAC and normal tissues. Bioinformatics analysis of transcriptomics and proteomics data evaluated genes associated with taurine metabolism. Identified taurine-associated gene was validated through gene modulation. Clinical implications were evaluated using patient data. Metabolomics analysis showed a 2.51-fold increase in taurine in PDAC compared to normal tissues (n=54). IHC confirmed this in independent samples (n=99 PDAC, 19 normal). Bioinformatics identified 2-aminoethanethiol dioxygenase (ADO) as a key gene modulating taurine metabolism. IHC on a tissue microarray (39 PDAC, 10 normal) confirmed elevated ADO in PDAC. The ADO-Taurine axis correlated with PDAC recurrence and disease-free survival. ADO knockdown reduced cancer cell proliferation and tumor growth in a mouse xenograft model. The MEK-related signaling pathway is suggested to be modulated by ADO-Taurine metabolism. Our multi-omics investigation revealed elevated taurine synthesis mediated by ADO upregulation in PDAC. The ADO-Taurine axis may serve as a biomarker for PDAC prognosis and a therapeutic target.
Collapse
Affiliation(s)
- Hoonsik Nam
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Woohyung Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Yun Ji Lee
- Department of Biomedical Sciences, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Jin-Mo Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung Hee Jung
- Department of Biomedical Sciences, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Song Cheol Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Sunghyouk Park
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
156
|
Xu R, Martinez-Bosch N, Rivera-Hueto F, Mulens-Arias V, Rubio-Moscardo F, Javier Conesa J, Navarro P, Vicente R, Rivera-Gil P. Validation of ZIP4 as a tumour-associated antigen for nanotargeting. J Drug Target 2025; 33:143-155. [PMID: 39283041 DOI: 10.1080/1061186x.2024.2405711] [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: 05/01/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 10/02/2024]
Abstract
Pancreatic ductal adenocarcinoma remains a highly aggressive and untreatable cancer. There is a need to develop a new PDAC-associated antigen-targeting drug delivery system to tackle this disease. We validated choosing ZIP4 as a putative target in PDAC theranostics. We developed a nanosystem composed of a fluorescent polystyrene core coated with gold nanoparticles onto which a ZIP4-specific polyclonal antibody is attached. The polystyrene core's fluorescence properties allow the nanosystem tracking by intravital imaging. We also developed two ZIP4-expressing cell lines by stably transfecting HEK293 and RWP1 cells with a ZIP4-coding plasmid that simultaneously provides cells with puromycin resistance. We studied the cell internalisation of the as-synthesised nanoparticles and demonstrated that ZIP4-expressing HEK293 and ZIP4-expressing RWP1 cells tended to take up more ZIP4-targeting nanoparticles. Moreover, we observed that ZIP4-targeting nanoparticles accumulated more in ZIP4-expressing HEK293 and RWP1 tumours when injected intravenously in a subcutaneous xenograft and an orthotopic in vivo model, respectively. Furthermore, the administration of these nanoparticles did not induce any significant systemic toxicity as determined by histological analysis of all organs. Altogether, these results provide the first evidence of the feasibility of using a ZIP4-targeting nanosystem further to design efficient therapeutic and diagnostic tools for PDAC.
Collapse
Affiliation(s)
- Ruixue Xu
- Integrative Biomedical Materials and Nanomedicine Lab, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| | - Neus Martinez-Bosch
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, Barcelona, Spain
| | | | - Vladimir Mulens-Arias
- Integrative Biomedical Materials and Nanomedicine Lab, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| | - Fanny Rubio-Moscardo
- Molecular Physiology Lab, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| | - J Javier Conesa
- Mistral Beamline, Experiment Division, ALBA Synchrotron (ALBA-CELLS), Barcelona, Spain
| | - Pilar Navarro
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, Barcelona, Spain
- Institute of Biomedical Research of Barcelona (IIBB)-CSIC, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rubén Vicente
- Molecular Physiology Lab, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| | - Pilar Rivera-Gil
- Integrative Biomedical Materials and Nanomedicine Lab, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, PRBB, Barcelona, Spain
| |
Collapse
|
157
|
James PD, Almousawi F, Salim M, Khan R, Tanuseputro P, Hsu AT, Coburn N, Alabdulkarim B, Talarico R, Gayowsky A, Webber C, Seow H, Sutradhar R. Development and Validation of a Survival Prediction Model for Patients With Pancreatic Cancer. Clin Transl Gastroenterol 2025; 16:e00774. [PMID: 39620578 PMCID: PMC11756872 DOI: 10.14309/ctg.0000000000000774] [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: 04/26/2024] [Accepted: 09/18/2024] [Indexed: 12/25/2024] Open
Abstract
INTRODUCTION Patients with pancreatic ductal adenocarcinoma (PDAC) face challenging treatment decisions following their diagnosis. We developed and validated a survival prognostication model using routinely available clinical information, patient-reported symptoms, performance status, and initial cancer-directed treatment. METHODS This retrospective cohort study included patients with PDAC from 2007 to 2020 using linked administrative databases in Ontario, Canada. Patients were randomly selected for model development (75%) and validation (25%). Using the development cohort, a multivariable Cox proportional hazards regression with backward stepwise variable selection was used to predict the probability of survival. Model performance was assessed on the validation cohort using the concordance index and calibration plots. RESULTS There were 17,450 patients (49% female) with a median age of 72 years (interquartile range 63-81) and a mean survival time of 9 months. In the derivation cohort, 1,469 patients (11%) had early stage, 4,202 (32%) had advanced stage disease, and 7,417 (57%) had unknown stage. The following factors were associated with an increased risk of death by more than 10%: tumor in the tail of the pancreas; advanced stage; hospitalization 3 months before diagnosis; congestive heart failure or dementia; low, moderate, or high pain score; moderate or high appetite score; high dyspnea and tiredness score; and a performance status score of 60-70 or lower. The calibration plot indicated good agreement with a C-index of 0.76. DISCUSSION This model accurately predicted one-year survival for PDAC using clinical factors, symptoms, and performance status. This model may foster shared decision making for patients and their providers.
Collapse
Affiliation(s)
- Paul D. James
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Gastroenterology, University Health Network, Toronto, Ontario, Canada
| | - Fatema Almousawi
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Gastroenterology, University Health Network, Toronto, Ontario, Canada
| | - Misbah Salim
- Division of Gastroenterology, University Health Network, Toronto, Ontario, Canada
| | - Rishad Khan
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Gastroenterology, University Health Network, Toronto, Ontario, Canada
| | - Peter Tanuseputro
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Bruyère Research Institute, Ottawa, Ontario, Canada
| | - Amy T. Hsu
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Bruyère Research Institute, Ottawa, Ontario, Canada
- Department of Family Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Natalie Coburn
- Division of General Surgery, University of Toronto, Toronto, Ontario, Canada
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Balqis Alabdulkarim
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Gastroenterology, University Health Network, Toronto, Ontario, Canada
| | - Robert Talarico
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | | | - Colleen Webber
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Hsien Seow
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Rinku Sutradhar
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Institute for Health Policy, Evaluation and Management, University of Toronto, Toronto, Ontario, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
158
|
Shahrokhi Nejad S, Razi S, Rezaei N. The role of AMPK in pancreatic cancer: from carcinogenesis to treatment. Clin Transl Oncol 2025; 27:70-82. [PMID: 38926257 DOI: 10.1007/s12094-024-03572-8] [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: 02/20/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Pancreatic cancer has doubled over the previous two decades. Routine therapies are becoming incredibly resistant and failing to compensate for the burden caused by this aggressive neoplasm. As genetic susceptibility has always been a highlighted concern for this disease, identifying the molecular pathways involved in the survival and function of pancreatic cancer cells provides insight into its variant etiologies, one of which is the role of AMPK. This regulating factor of cell metabolism is crucial in the homeostasis and growth of the cell. Herein, we review the possible role of AMPK in pancreatic cancer while considering its leading effects on glycolysis and autophagy. Then, we assess the probable therapeutic agents that have resulted from the suggested pathways. Studying the underlying genetic changes in pancreatic cancer provides a chance to detect and treat patients suffering from advanced stages of the disease, and those who have given up their hope on conventional therapies can gain an opportunity to combat this cancer.
Collapse
Affiliation(s)
- Shahrzad Shahrokhi Nejad
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, 14194, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, 14194, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
159
|
Hung YS, Chiu TJ, Chen YY, Chou WC. Validation of a Prognostic Nomogram for Patients with Metastatic Pancreatic Cancer Treated with Nanoliposomal Irinotecan as Second-Line Therapy. Cancer Control 2025; 32:10732748251333040. [PMID: 40216411 PMCID: PMC12033549 DOI: 10.1177/10732748251333040] [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: 12/20/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
IntroductionNanoliposomal irinotecan (nal-IRI) plus 5-fluorouracil and leucovorin (5-FU/LV) is an established second-line therapy for metastatic pancreatic ductal adenocarcinoma (PDAC). We previously developed a prognostic model (CGMH nomogram) to predict overall survival (OS) in patients receiving second-line chemotherapy before the nal-IRI + 5-FU/LV era. Herein, we aimed to validate the CGMH nomogram in a real-world cohort treated with nal-IRI plus 5-FU/LV, the current standard second-line treatment for metastatic PDAC.MethodsA retrospective cohort of 148 patients with metastatic PDAC treated with second-line nal-IRI + 5-FU/LV was analyzed. Prognostic scores were assigned using the CGMH nomogram, with patients stratified into tertiles as good, intermediate, and poor prognostic groups. Predictive performance was assessed using the concordance index (c-index) and calibration plots.ResultsOur cohort had a median OS of 6.1 months. Patients in the good, intermediate, and poor prognostic groups had median OS of 8.7 (95% confidence interval [CI], 6.7-10.7), 5.7 (95% CI, 5.3-6.3), and 4.0 (95% CI, 2.8-5.2) months, respectively. Compared with the good group, intermediate and poor groups had hazard ratios of 1.99 (95% CI, 1.29-3.07, P = .002) and 3.18 (95% CI, 1.87-5.40, P < .001), respectively. The nomogram demonstrated strong predictive ability, with c-indices of 0.73 and 0.70 for 6- and 12-month OS predictions, respectively. Calibration plots displayed excellent agreement between predicted and observed survival.ConclusionThe CGMH nomogram reliably predicted survival outcomes in nal-IRI + 5-FU/LV-treated patients with metastatic PDAC, and validation supported its use in clinical decision-making and personalized treatment planning.
Collapse
Affiliation(s)
- Yu-Shin Hung
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyaun, Taiwan
| | - Tai-Jan Chiu
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Kaohsiung, Chang Gung University College of Medicine, Taoyaun, Taiwan
| | - Yen-Yang Chen
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Kaohsiung, Chang Gung University College of Medicine, Taoyaun, Taiwan
| | - Wen-Chi Chou
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyaun, Taiwan
| |
Collapse
|
160
|
Ariake K, Mizuma M, Unno M, Satoi S, Yamamoto N, Hayashi M, Kawai M, Akita H, Toyoda E, Fujii T, Sasaki M, Hakamada K, Watanabe J, Hatano E, Hidaka M, Hirano S, Kurahara H, Matsumoto I, Honda G, Ogura T, Nakamura M, Endo I. Optimal treatment strategy for patients with pancreatic cancer having positive peritoneal cytology: A nationwide multicenter retrospective cohort study supervised by the Japanese Society of Hepato-Biliary-Pancreatic Surgery. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2025; 32:69-81. [PMID: 39317950 PMCID: PMC11780303 DOI: 10.1002/jhbp.12074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
BACKGROUND The aim of this study was to determine the optimal treatment for patients with pancreatic cancer (PaCa) having positive peritoneal cytology (PPC). METHODS This multicenter retrospective study included patients with PPC treated at 78 high-volume centers between January 2012 and December 2020. Prognoses after resection (S-group) and initiation of nonsurgical treatment (N-group) were compared. Prognostic factors for survival in both groups were analyzed. Detailed characteristics of conversion surgery (CS) in the N-group were evaluated. RESULTS In total, 568 enrolled patients were classified into an S-group (n = 445) or an N-group (n = 123). Median survival times (MSTs) were 19.0 months and 19.3 months, respectively, with no significant difference in prognosis (p = .845). The intervenable prognostic factors for survival were adjuvant treatment in the S-group (p < .001) and CS in the N-group (p < .001). Following CS, the MST was prolonged to 45.6 months, and peritoneal or liver recurrence decreased considerably. CS can be expected if PPC is diagnosed before neoadjuvant treatment and when combination treatment is initiated. CONCLUSION Surgical resection may not be beneficial for improving survival when PPC is evident. Chemotherapy aiming for CS may be the optimal treatment for such patients.
Collapse
Affiliation(s)
- Kyohei Ariake
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
- Department of Gastroenterological SurgerySendai City Medical Center Sendai Open HospitalSendaiJapan
| | - Masamichi Mizuma
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Michiaki Unno
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Sohei Satoi
- Department of SurgeryKansai Medical UniversityHirakataJapan
| | - Naoto Yamamoto
- Department of Gastrointestinal SurgeryKanagawa Cancer CenterYokohamaJapan
| | - Masamichi Hayashi
- Department of Surgery, Graduate School of MedicineNagoya UniversityNagoyaJapan
| | - Manabu Kawai
- Second Department of SurgeryWakayama Medical University School of MedicineWakayamaJapan
| | - Hirofumi Akita
- Department of Gastroenterological SurgeryOsaka International Cancer InstituteOsakaJapan
| | - Eiji Toyoda
- Department of SurgeryOtsu Red Cross HospitalOtsuJapan
| | - Tsutomu Fujii
- Department of Surgery and Science, Faculty of MedicineAcademic Assembly, University of ToyamaToyamaJapan
| | - Masaru Sasaki
- Department of SurgeryJA Hiroshima General HospitalHatsukaichiJapan
| | - Kenichi Hakamada
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| | - Jota Watanabe
- Department of Gastrointestinal SurgeryEhime Prefectural Central HospitalMatsuyamaJapan
| | - Etsuro Hatano
- Department of SurgeryGraduate School of Medicine, Kyoto UniversityKyotoJapan
| | - Masaaki Hidaka
- Department of Digestive and General SurgeryShimane University Faculty of MedicineIzumoJapan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery IIHokkaido University Faculty of MedicineSapporoJapan
| | - Hiroshi Kurahara
- Department of Digestive SurgeryKagoshima UniversityKagoshimaJapan
| | - Ippei Matsumoto
- Department of Surgery, Faculty of MedicineKindai UniversityOsakasayamaJapan
| | - Goro Honda
- Department of SurgeryInstitute of Gastroenterology, Tokyo Women's Medical UniversityTokyoJapan
| | - Toshiro Ogura
- Department of Gastroenterological SurgerySaitama Cancer CenterSaitamaJapan
| | - Masafumi Nakamura
- Department of Surgery and OncologyGraduate School of Medical Sciences, Kyushu UniversityFukuokaJapan
- Japanese Society of Hepato‐Biliary‐Pancreatic SurgeryTokyoJapan
| | - Itaru Endo
- Japanese Society of Hepato‐Biliary‐Pancreatic SurgeryTokyoJapan
- Department of Gastroenterological Surgery, Graduate School of MedicineYokohama City UniversityYokohamaJapan
| |
Collapse
|
161
|
Cockrum P, Dennen S, Brown A, Briggs J, Paluri R. Real-world clinical outcomes and economic burden of metastatic pancreatic ductal adenocarcinoma: a systematic review. Future Oncol 2025; 21:241-260. [PMID: 39648649 PMCID: PMC11792790 DOI: 10.1080/14796694.2024.2435253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/25/2024] [Indexed: 12/10/2024] Open
Abstract
AIMS This systematic review summarizes real-world clinical outcomes and economic burden of first-line FOLFIRINOX (FFX)/modified FFX (mFFX) and nab-paclitaxel plus gemcitabine (GnP) in metastatic pancreatic ductal adenocarcinoma in the US. METHODS Embase and MEDLINE were searched for materials published since 2014; citations were reviewed in a two-step process. Included studies were qualitatively synthesized. RESULTS Searches yielded 2,528 citations; 29 were included (17 clinical studies/12 economic studies). In 9/17 clinical studies, median overall survival (mOS) ranged from 4.7 months to 11.4 months for FFX/mFFX, with the unweighted median of the estimates within this range being 9.2 months; for GnP mOS ranged from 3.6 to 9.8 months, and the unweighted median of the estimates was 6.9 months. In 8/17 studies, grade 3/4 anemia, neutropenia, and thrombocytopenia were the most commonly reported adverse events. Across economic burden studies, total costs were similar between the 2 groups. Outpatient, supportive care, and granulocyte colony-stimulating factor costs were higher for the FFX generic regimen, and chemotherapy costs were higher for the GnP branded regimen. CONCLUSIONS Real-world OS in FFX- and GnP-treated populations was shorter than that in clinical trials, and total costs of FFX and GnP were similar, but with differences in cost components.
Collapse
Affiliation(s)
- Paul Cockrum
- Medical Affairs, Ipsen Biopharmaceuticals Inc, Cambridge, MA, USA
| | | | | | | | - Ravi Paluri
- Hematology and Oncology, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| |
Collapse
|
162
|
Gayibov E, Sychra T, Spálenková A, Souček P, Oliverius M. The use of patient-derived xenografts and patient-derived organoids in the search for new therapeutic regimens for pancreatic carcinoma. A review. Biomed Pharmacother 2025; 182:117750. [PMID: 39689516 DOI: 10.1016/j.biopha.2024.117750] [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: 09/06/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/19/2024] Open
Abstract
Patient-derived organoids (PDOs) and xenografts (PDXs) are powerful tools for personalized medicine in pancreatic cancer (PC) research. This study explores the complementary strengths of PDOs and PDXs in terms of practicality, genetic fidelity, cost, and labor considerations. Among other models like 2D cell cultures, spheroids, cancer-on-chip systems, cell line-derived xenografts (CDX), and genetically engineered mouse models (GEMMs), PDOs and PDXs uniquely balance genetic fidelity and personalized medicine potential, offering distinct advantages over the simplicity of 2D cultures and the advanced, but often resource-intensive, GEMMs and cancer-on-chip systems. PDOs excel in high-throughput drug screening due to their ease of use, lower cost, and shorter experimental timelines. However, they lack a complete tumor microenvironment. Conversely, PDXs offer a more complex microenvironment that closely reflects patient tumors, potentially leading to more clinically relevant results. Despite limitations in size, number of specimens, and engraftment success, PDXs demonstrate significant concordance with patient responses to treatment, highlighting their value in personalized medicine. Both models exhibit significant genetic fidelity, making them suitable for drug sensitivity testing. The choice between PDOs and PDXs depends on the research focus, resource availability, and desired level of microenvironment complexity. PDOs are advantageous for high-throughput screening of a diverse array of potential therapeutic agents due to their relative ease of culture and scalability. PDXs, on the other hand, offer a more physiologically relevant model, allowing for a comprehensive evaluation of drug efficacy and mechanisms of action.
Collapse
Affiliation(s)
- Emin Gayibov
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomáš Sychra
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic; Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Department of General Surgery, 3rd Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Alžběta Spálenková
- Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Souček
- Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| | - Martin Oliverius
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic; Department of General Surgery, 3rd Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic.
| |
Collapse
|
163
|
Stefàno E, Rovito G, Cossa LG, Castro FD, Vergaro V, Ali A, My G, Migoni D, Muscella A, Marsigliante S, Benedetti M, Fanizzi FP. Novel Pt (II) Complexes With Anticancer Activity Against Pancreatic Ductal Adenocarcinoma Cells. Bioinorg Chem Appl 2024; 2024:5588491. [PMID: 39886428 PMCID: PMC11779987 DOI: 10.1155/bca/5588491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/16/2024] [Indexed: 02/01/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive type of solid tumor that is becoming more common. cis-[PtCl2 (NH3)2] (in short cisplatin or CDDP) has been shown to be effective in treating various cancers, including PDAC. However, the development of resistance to chemotherapy drugs has created a need for the synthesis of new anticancer agents. Platinum-based drugs containing the bidentate ligand phenanthroline have been found to have strong antitumor activity due to their ability to cause DNA damage. In this study, we examined the ability of two Pt (II) cationic complexes, [Pt(η 1-C2H4OR) (DMSO) (phen)]+ (in short Pt-EtORSOphen; R = Me, 1; Et, 2), to inhibit the growth and spread of BxPC-3 PDAC cells, in comparison to CDDP. The length of the alkyl chain and its associated lipophilic properties did not affect the anticancer effects of complexes 1 and 2 in BxPC-3 cells. However, it did appear to influence the rapid loss of mitochondrial membrane potential (ΔΨM), suggesting that these complexes could potentially be used as mitochondria-targeted lipophilic cations in anticancer therapy.
Collapse
Affiliation(s)
- Erika Stefàno
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Gianluca Rovito
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Luca G. Cossa
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Federica De Castro
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Viviana Vergaro
- Department of Experimental Medicine, University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Asjad Ali
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Giulia My
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Danilo Migoni
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Antonella Muscella
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Santo Marsigliante
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Michele Benedetti
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Francesco Paolo Fanizzi
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| |
Collapse
|
164
|
Finan JM, Di Niro R, Park SY, Jeong KJ, Hedberg MD, Smith A, McCarthy GA, Haber AO, Muschler J, Sears RC, Mills GB, Gmeiner WH, Brody JR. The polymeric fluoropyrimidine CF10 overcomes limitations of 5-FU in pancreatic ductal adenocarcinoma cells through increased replication stress. Cancer Biol Ther 2024; 25:2421584. [PMID: 39513592 PMCID: PMC11552260 DOI: 10.1080/15384047.2024.2421584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease soon to become the second leading cause of cancer deaths in the US. Beside surgery, current therapies have narrow clinical benefits with systemic toxicities. FOLFIRINOX is the current standard of care, one component of which is 5- Fluorouracil (5-FU), which causes serious gastrointestinal and hematopoietic toxicities and is vulnerable to resistance mechanisms. Recently, we have developed polymeric fluoropyrimidines (F10, CF10) which unlike 5-FU, are, in principle, completely converted to the thymidylate synthase inhibitory metabolite FdUMP, without generating appreciable levels of ribonucleotides that cause systemic toxicities while displaying much stronger anti-cancer activity. Here, we confirm the potency of CF10 and investigate enhancement of its efficacy through combination with inhibitors in vitro targeting replication stress, a hallmark of PDAC cells. CF10 is 308-times more potent as a single agent than 5-FU and was effective in the nM range in primary patient derived models. Further, we find that activity of CF10, but not 5-FU, is enhanced through combination with inhibitors of ATR and Wee1 that regulate the S and G2 DNA damage checkpoints and can be reversed by addition of dNTPs indicative of CF10 acting, at least in part, through inducing replication stress. Our results indicate CF10 has the potential to supersede the established benefit of 5-FU in PDAC treatment and indicate novel combination approaches that should be validated in vivo and may be beneficial in established regimens that include 5-FU.
Collapse
Affiliation(s)
- Jennifer M. Finan
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
| | - Roberto Di Niro
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Soon Young Park
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Kang Jin Jeong
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Madeline D. Hedberg
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Alexander Smith
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Grace A. McCarthy
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
| | - Alex O. Haber
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - John Muschler
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Rosalie C. Sears
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Gordon B. Mills
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Development and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
| | - William H. Gmeiner
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jonathan R. Brody
- Department of Surgery, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
165
|
Zeng J, Wang C, Ruge F, Ji EK, Martin TA, Sanders AJ, Jia S, Hao C, Jiang WG. EPLIN, a prospective oncogenic molecule with contribution to growth, migration and drug resistance in pancreatic cancer. Sci Rep 2024; 14:30850. [PMID: 39730634 DOI: 10.1038/s41598-024-81485-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: 03/26/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024] Open
Abstract
Most pancreatic cancer patients are diagnosed at advanced stages, with poor survival rates and drug resistance making pancreatic cancer one of the highest causes of cancer death in the UK. Understanding the underlying mechanism behind its carcinogenesis, metastasis and drug resistance has become an essential task for researchers. We have discovered that a well-established tumour suppressor, EPLIN, has an oncogenic rather than suppressive role in pancreatic cancer. Notably, upregulation of EPLIN was observed in pancreatic cancer samples compared to normal samples at RNA and protein levels. Moreover, the presence of EPLIN resulted in poor clinical outcomes in patients. We also report that inhibition of EPLIN led to reduced cellular growth and migration in pancreatic cancer cells. EPLIN regulates expression and phosphorylation levels of several key players in MAPK and PIK3CA-AKT signalling pathways, as well as key contributors of EMT. Furthermore, EPLIN mediates the inhibitory ability PIK3 kinases, MEK and ERK inhibitors have on cell migration. EPLIN was also found to have an impact on pancreatic cancer cells response to chemotherapeutic and EGFR/HER2 targeted therapeutic agents, namely gemcitabine, fluorouracil (5FU) and neratinib (Nerlynx).
Collapse
Affiliation(s)
- Jianyuan Zeng
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff, CF14 4XN, UK
| | - Cai Wang
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff, CF14 4XN, UK
| | - Fiona Ruge
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff, CF14 4XN, UK
| | - Edison Ke Ji
- Gastrointestinal Cancer Centre, Peking University Cancer Hospital, Peking University, Fucheng Road, Haidian District, Beijing, China
| | - Tracey A Martin
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff, CF14 4XN, UK
| | - Andrew J Sanders
- School of Education and Science, University of Gloucestershire, Francis Close Hall, Swindon Road, Cheltenham, GL50 4AZ, UK
| | - Shuqin Jia
- Gastrointestinal Cancer Centre, Peking University Cancer Hospital, Peking University, Fucheng Road, Haidian District, Beijing, China
| | - Chunyi Hao
- Gastrointestinal Cancer Centre, Peking University Cancer Hospital, Peking University, Fucheng Road, Haidian District, Beijing, China
| | - Wen G Jiang
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff, CF14 4XN, UK.
| |
Collapse
|
166
|
Tan JZC, Zhang Z, Goh HX, Ngeow J. BRCA and Beyond: Impact on Therapeutic Choices Across Cancer. Cancers (Basel) 2024; 17:8. [PMID: 39796639 PMCID: PMC11718952 DOI: 10.3390/cancers17010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Identifying patients with gBRCAm is crucial to facilitate screening strategies, preventive measures and the usage of targeted therapeutics in their management. This review examines the evidence for the latest predictive and therapeutic approaches in BRCA-associated cancers. CLINICAL DESCRIPTION Data supports the use of adjuvant olaparib in patients with gBRCAm high-risk HER2-negative breast cancer. In advanced gBRCAm HER2-negative breast cancer, the PARPis talazoparib and olaparib have demonstrated benefit over standard chemotherapy. In ovarian cancer, olaparib, niraparib or rucaparib can be used as monotherapy in frontline maintenance. Olaparib and bevacizumab as a combination can also be used as frontline maintenance. In the relapsed platinum-sensitive setting, olaparib, niraparib and rucaparib are effective maintenance options in BRCAm patients who are PARPi naive. Both olaparib and rucaparib are effective options in BRCAm metastatic castrate-resistant prostate cancer (mCRPC). Evidence also exists for the benefit of PARPi combinations in mCRPC. In metastatic pancreatic cancer, olaparib can be used in gBRCAm patients who are responding to platinum chemotherapy. However, there may be a development of PARPi resistance. Understanding the pathophysiology that contributes to such resistance may allow the development of novel therapeutics. Combination therapy appears to have promising results in emerging trials. Seeking avenues for subsidised genetic testing can reduce the total costs of cancer management, leading to improve detection rates. CONCLUSION Identifying breast, ovarian, pancreatic and prostate cancer patients with gBRCAm plays a crucial predictive role in selecting those who will benefit significantly from PARPi therapy. The use of PARPi in gBRCAm HBOC-related cancers has resulted in significant survival benefits. Beyond BRCA1/2, HRR gene assessment and the consideration of other cancer predisposition syndromes may allow more patients to be eligible for and benefit from targeted therapies.
Collapse
Affiliation(s)
- Joshua Zhi Chien Tan
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore 168583, Singapore; (J.Z.C.T.); (Z.Z.)
| | - Zewen Zhang
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore 168583, Singapore; (J.Z.C.T.); (Z.Z.)
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore 168583, Singapore
| | - Hui Xuan Goh
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore 168583, Singapore
| | - Joanne Ngeow
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore 168583, Singapore; (J.Z.C.T.); (Z.Z.)
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore 168583, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 11 Mandalay Rd, Singapore 308232, Singapore
| |
Collapse
|
167
|
Lee TS. Are Probiotics Beneficial or Harmful for Pancreatic Cancer Outcomes? Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10437-7. [PMID: 39714574 DOI: 10.1007/s12602-024-10437-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Pancreatic cancer is influenced by interactions between cancer cells and the tumor microenvironment (TME), including tumor-infiltrating lymphocytes (TILs). Specifically, CD8 + T cells impact prognosis by eliminating cancer cells. Recent studies have revealed that microbiomes are present in pancreatic tissues and may affect tumor growth and immune responses. Additionally, recent studies revealed that the abundance of Bacteroides, Lactobacillus, and Peptoniphilus are associated with poor pancreatic cancer prognosis. This study investigates the role of oral probiotics in influencing pancreatic cancer outcomes. We retrospectively reviewed patients aged ≥ 18 years with pathologically confirmed pancreatic cancer from Seoul National University Hospital between January 2011 and January 2023. We investigated progression-free survival and overall survival between the control group and the probiotics group. Among pancreatic cancer patients undergoing palliative chemotherapy without radiotherapy and resection, there was a significant difference in overall survival (OS) when comparing the control group to the probiotics group (median: 10 months (9-11) vs. 12 months (9-19), p = 0.026). Regardless of the type of probiotics, oral probiotics may have a positive impact, but further research is still needed to understand the underlying immunological mechanisms.
Collapse
Affiliation(s)
- Tae Seung Lee
- Division of Gastroenterology, Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, 101, Daehak-Ro, Jongno-Gu, Seoul, Republic of Korea.
| |
Collapse
|
168
|
Maiti A, Mondal S, Choudhury S, Bandopadhyay A, Mukherjee S, Sikdar N. Oncometabolites in pancreatic cancer: Strategies and its implications. World J Exp Med 2024; 14:96005. [PMID: 39713078 PMCID: PMC11551704 DOI: 10.5493/wjem.v14.i4.96005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/24/2024] [Accepted: 09/14/2024] [Indexed: 10/31/2024] Open
Abstract
Pancreatic cancer (PanCa) is a catastrophic disease, being third lethal in both the genders around the globe. The possible reasons are extreme disease invasiveness, highly fibrotic and desmoplastic stroma, dearth of confirmatory diagnostic approaches and resistance to chemotherapeutics. This inimitable tumor microenvironment (TME) or desmoplasia with excessive extracellular matrix accumulation, create an extremely hypovascular, hypoxic and nutrient-deficient zone inside the tumor. To survive, grow and proliferate in such tough TME, pancreatic tumor and stromal cells transform their metabolism. Transformed glucose, glutamine, fat, nucleotide metabolism and inter-metabolite communication between tumor and TME in synergism, impart therapy resistance, and immunosuppression in PanCa. Thus, a finer knowledge of altered metabolism would uncover its metabolic susceptibilities. These unique metabolic targets may help to device novel diagnostic/prognostic markers and therapeutic strategies for better management of PanCa. In this review, we sum up reshaped metabolic pathways in PanCa to formulate detection and remedial strategies of this devastating disease.
Collapse
Affiliation(s)
- Arunima Maiti
- Suraksha Diagnostics Pvt Ltd, Newtown, Rajarhat, Kolkata 700156, West Bengal, India
| | - Susmita Mondal
- Department of Zoology, Diamond Harbour Women’s University, Diamond Harbour 743368, West Bengal, India
| | - Sounetra Choudhury
- Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, West Bengal, India
| | | | - Sanghamitra Mukherjee
- Department of Pathology, RG Kar Medical College and Hospital, Kolkata 700004, West Bengal, India
| | - Nilabja Sikdar
- Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, West Bengal, India
- Scientist G, Estuarine and Coastal Studies Foundation, Howrah 711101, West Bengal, India
| |
Collapse
|
169
|
Xiang Z, Ma L, Li Z, Fu Y, Pan Y. Cost-effectiveness analysis of first-line combination chemotherapy regimens for metastatic pancreatic cancer and evidence-based pricing strategy of liposomal irinotecan in China. Front Pharmacol 2024; 15:1488645. [PMID: 39759454 PMCID: PMC11695189 DOI: 10.3389/fphar.2024.1488645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Background The phase III NAPOLI-3 trial, which upgraded FOLFIRINOX (leucovorin, fluorouracil, irinotecan and oxaliplatin) to NALIRIFOX (liposomal irinotecan, oxaliplatin, leucovorin, and fluorouracil), demonstrated the superiority of NALIRIFOX over GEMNABP (gemcitabine and nab-paclitaxel) as the first-line treatment for metastatic pancreatic ductal adenocarcinoma. The purpose of this study was to assess the cost-effectiveness of NALIRIFOX, FOLFIRINOX, and GEMNABP, and to simulate the price of liposomal irinotecan at which NALIRIFOX could achieve cost-effectiveness. Methods A partitioned survival model was performed to evaluate the cost-effectiveness of NALIRIFOX, FOLFIRINOX and GEMNABP from the perspective of the Chinese healthcare system. Survival data was obtained from a recently published network meta-analysis (NMA). Drug prices were collected from the database of the Hunan Province Drug and Medical Consumables Procurement Management Subsystem. Other cost and utility values were sourced from established literature. Cumulative costs, LYs (life-years), quality-adjusted life years (QALYs), incremental cost-effectiveness ratios (ICERs), net monetary benefits (NMBs) and incremental net monetary benefits (INMBs) were the main outputs. Furthermore, the variations in ICER were analyzed as the price of liposomal irinotecan gradually decreased when comparing NALIRIFOX with FOLFIRINOX or GEMNABP. The robustness of the model was assessed by sensitivity analysis and scenario analysis. Results At the willingness-to-pay (WTP) threshold of $38,223.34, GEMNABP was the favored treatment. NALIRIFOX was associated with the highest LYs, QALYs, and cost. The cost-effectiveness of NALIRIFOX would be obtained if the price of liposomal irinotecan was less than $3.36/mg and $2.08/mg compared to FOLFIRINOX and GEMNABP, respectively, without considering the patient assistance program (PAP). Sensitivity analysis and scenario analysis revealed that the results of the model were stable. Conclusion From an economic standpoint, GEMNABP represents the favored choice in the prevailing market conditions among these three first-line combination chemotherapy regimens. The price simulation of liposomal irinotecan conducted in this study could provide valuable evidence for healthcare decision-making. Further evidence regarding the budget impact is still needed.
Collapse
Affiliation(s)
- Zuojuan Xiang
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Ling Ma
- Department of Clinical pharmacy, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Zhengxiong Li
- School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, China
| | - Yingzhou Fu
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Yong Pan
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| |
Collapse
|
170
|
Pham TD, Becker JH, Metropulos AE, Mubin N, Spaulding C, Bentrem DJ, Munshi HG. Regorafenib induces DNA damage and enhances PARP inhibitor efficacy in pancreatic ductal carcinoma. BMC Cancer 2024; 24:1562. [PMID: 39707244 DOI: 10.1186/s12885-024-13334-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: 04/27/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND There is increasing interest in enhancing the response of the PARP inhibitor olaparib, which is currently approved for pancreatic ductal adenocarcinoma (PDAC) patients with defects in DNA damage repair associated with germline BRCA1/2 mutations. Moreover, agents that can mimic these defects in the absence of germline BRCA1/2 mutations are an area of active research in hopes of increasing the number of patients eligible for treatment with PARP inhibitors. The extent to which regorafenib, an FDA-approved tyrosine kinase inhibitor, can be used to enhance the efficacy of PARP inhibitors in PDAC cells without known BRCA1/2 mutations remains to be investigated. METHODS Comet assay, cell cycle analysis, western blotting, and immunofluorescent detection of H2AXS139 were used to evaluate the extent to which regorafenib induces DNA damage in PDAC cell lines. The effects of regorafenib, either alone or in combination with PARPi inhibitors, on PDAC cell death were assessed by Annexin V/PI co-staining assay in cell lines and by immunohistochemistry staining for cleaved caspase-3 in mouse tumors and in ex vivo slice cultures of human PDAC tumors. Flow cytometry-based analysis was used to evaluate the ability of regorafenib to reprogram PDAC tumor microenvironment. RESULTS We now show that regorafenib, a tyrosine-kinase inhibitor with efficacy in several gastrointestinal malignancies, can enhance the response of olaparib in pancreatic cancer. While regorafenib induces DNA damage and limits the ability of PDAC cells to resolve the damage, regorafenib by itself does not induce apoptosis. However, regorafenib in combination with olaparib further induces DNA damage in vitro, in tumor-bearing mice, and in ex vivo slice cultures of human PDAC tumors, resulting in increased apoptosis compared to olaparib alone. Notably, we show that the efficacy of the combination treatment is not dependent on cytolytic T cells. CONCLUSIONS Together, these findings demonstrate that regorafenib can attenuate DNA damage response and potentiate the efficacy of PARP inhibitors in PDAC tumors.
Collapse
Affiliation(s)
- Thao D Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.
| | - Jeffrey H Becker
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Anastasia E Metropulos
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Nida Mubin
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Christina Spaulding
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - David J Bentrem
- Jesse Brown VA Medical Center, Chicago, IL, USA
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hidayatullah G Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Jesse Brown VA Medical Center, Chicago, IL, USA.
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.
| |
Collapse
|
171
|
Wahida A, George B, Kurzrock R. At the right time: Moving precision therapy to newly diagnosed cancer. MED 2024; 5:1463-1465. [PMID: 39674173 DOI: 10.1016/j.medj.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/04/2024] [Accepted: 10/18/2024] [Indexed: 12/16/2024]
Abstract
Precision oncology aims to match the right drug(s) to the right patient. Equally important is ensuring that precision therapies are offered at the right time. Transformative, rather than incremental, outcome improvement may require treatment at diagnosis rather than in the advanced/metastatic setting after genomic evolution.
Collapse
Affiliation(s)
- Adam Wahida
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Ben George
- MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Razelle Kurzrock
- MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA; WIN Consortium, Paris, France.
| |
Collapse
|
172
|
Liu Q, Li R, Zhu W, Zheng P. Case report: Microsatellite instability-high pancreas adenosquamous carcinoma with postoperative liver metastasis recurrence treated with multimodality therapy achieving complete pathological response. Front Immunol 2024; 15:1456343. [PMID: 39726603 PMCID: PMC11669589 DOI: 10.3389/fimmu.2024.1456343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Pancreatic adenosquamous carcinoma (PASC) is a rare subtype of pancreatic cancer (PC), with no established consensus on the optimal treatment for postoperative liver metastasis recurrence. We report a case of a 48-year-old male patient who underwent radical surgery and was pathologically diagnosed with microsatellite instability-high (MSI-H) PASC. The patient experienced liver metastasis recurrence following single-agent gemcitabine adjuvant chemotherapy. After one session of transarterial chemoembolization (TACE) with oxaliplatin, fluorouracil, and epirubicin, followed by six cycles of adjuvant chemotherapy with gemcitabine and nab-paclitaxel combined with sintilimab immunotherapy and bevacizumab targeted therapy, complete pathological regression of the liver metastasis was achieved. The patient has now reached a 24-month survival period and continues to be monitored at our center. This case illustrates the promise of the proposed treatment regimen, highlighting the significant potential of multimodality strategies in managing metastatic recurrence of MSI-H PASC.
Collapse
Affiliation(s)
- Qinghua Liu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Ruoyun Li
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Wei Zhu
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, China
| | - Pengfei Zheng
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
173
|
Wang D, Wang X, Li Y, Wang X, Wang X, Su J, Wang A, Lv K, Liu M, Xia G. Improved Antitumor Efficiency of N4 -Tetradecyloxycarbonyl Gemcitabine-Loaded Liposomes for Pancreatic Cancer Chemotherapy. Int J Nanomedicine 2024; 19:13391-13410. [PMID: 39679246 PMCID: PMC11646436 DOI: 10.2147/ijn.s485861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/23/2024] [Indexed: 12/17/2024] Open
Abstract
Background Gemcitabine (Gem) is one of the first-line chemotherapy drugs for pancreatic cancer treatment. However, its short half-life in plasma and adverse effects limited its broader application. Methods A novel Gem derivative (N4 -tetradecyloxycarbonyl gemcitabine, tcGem) was synthesized and encapsulated into liposomes (LipotcGem) to overcome the above shortcomings. Results LipotcGem has been successfully formulated, with the average size of 115 nm, zeta potential values of -36 mV, encapsulation efficiency of up to 98%, and drug loading capacity of 8.1%. Compared to Gem, LipotcGem improved in vitro antitumor activity significantly, as evidenced by the lower IC50, the higher percentage of apoptotic cells, the stronger ability to inhibit cell migration and invasion due to the higher cellular accumulation (100 times). Additionally, the endocytosis of LipotcGem was mainly mediated by caveolae, and was then processed in the lysosome, where tcGem was released and hydrolyzed into Gem. LipotcGem inhibited tumor growth by 70% in subcutaneous xenograft model and 90% in orthotopic xenograft model, respectively. LipotcGem suppressed tumor metastasis and prolonged survival without perceptible systemic toxicity, which may be caused by the longer t1/2 in vivo (3.5 times, 5.23 vs 1.46 h) and more enrichment in tumor tissue (750 times). Conclusion LipotcGem significantly increased the anti-tumor efficiency and decreased the toxicity for chemotherapy of pancreatic cancer.
Collapse
Affiliation(s)
- Dan Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Xiaobo Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Yan Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Xiaowei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Xuelei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Jiayi Su
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Apeng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Kai Lv
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Mingliang Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Guimin Xia
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| |
Collapse
|
174
|
Wu JW, Zhou CF, Han ZX, Zhang H, Yan J, Chen J, Wang CB, Qin ZQ, Mao Y, Tang XY, Zhu LJ, Wei XW, Cui DH, Yang XL, Shi M, Zhao LQ, Jiang JL, Zhu WY, Wang HM, Wang C, Zhu LJ, Zhang J. Anlotinib plus chemotherapy as a first-line treatment for gastrointestinal cancer patients with unresectable liver metastases: a multicohort, multicenter, exploratory trial. Signal Transduct Target Ther 2024; 9:344. [PMID: 39648217 PMCID: PMC11625826 DOI: 10.1038/s41392-024-02051-4] [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: 07/31/2024] [Revised: 10/26/2024] [Accepted: 11/04/2024] [Indexed: 12/10/2024] Open
Abstract
This multicohort phase II trial (ALTER-G-001; NCT05262335) aimed to assess the efficacy of first-line anlotinib plus chemotherapy for gastrointestinal (GI) cancer patients with unresectable liver metastases. Eligible patients with colorectal cancer (Cohort A) or noncolorectal and nonesophageal GI cancer (Cohort C) received six cycles of anlotinib plus standard chemotherapeutic regimens followed by anlotinib plus metronomic capecitabine as a maintenance therapy. Liver metastasectomy can be performed when liver metastases are converted to resectable lesions. The primary outcome was the investigator-confirmed objective response rate (ORR) in the intention-to-treat population. Among the 47 patients in Cohort A, the ORR was 40.4% (95% CI 26.4-55.7), including 1 with a complete response (CR) and 18 who achieved a partial response (PR). The median progression-free survival (PFS) was 8.7 months (95% CI 7.3-NE), and the median overall survival (OS) was not reached. In Cohort C, 14 of 44 patients achieved a PR, with an ORR of 31.8% (95% CI 18.6-47.6). The PFS and OS were 5.8 months (95% CI 4.8-6.5) and 11.4 months (95% CI 5.8-19.3), respectively. The liver metastasectomy rate in patients with liver-limited disease was 22.7% (5/22) in Cohort A and 6.7% (2/30) in Cohort C. For pancreatic cancer patients, the ORR of the efficacy-evaluable population was 36.0% (9/25), and those with liver-limited metastasis had better survival. Moreover, no new safety concerns emerged. In conclusion, an anlotinib-based first-line regimen demonstrated promising antitumor activity among GI cancer patients with unresectable liver metastases and led to liver metastasectomy in selected patients.
Collapse
Affiliation(s)
- Jun-Wei Wu
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen-Fei Zhou
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng-Xiang Han
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Yan
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jun Chen
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Chun-Bin Wang
- Department of Oncology, The Third People's Hospital of Yancheng, Yancheng, China
| | - Zhi-Quan Qin
- Department of Medical Oncology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou, China
| | - Yong Mao
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xin-Yu Tang
- Department of Oncology, Wuxi Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Wuxi, China
| | - Liang-Jun Zhu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Nanjing, China
| | - Xiao-Wei Wei
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Dong-Hai Cui
- Department of Internal Medicine, Anyang Tumor Hospital, Anyang, China
| | - Xiu-Li Yang
- Department of Oncology, First Affiliated Hospital of Nanyang Medical College, Nanyang, China
| | - Min Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Qin Zhao
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin-Ling Jiang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-You Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hong-Mei Wang
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chun Wang
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Ling-Jun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
175
|
Mizutani S, Taniai N, Sukegawa M, Haruna T, Furuki H, Takata H, Ueda J, Yoshioka M, Aimoto T, Sakamoto S, Suzuki K, Nakamura Y, Yoshida H. Pancreatectomy with Celiac Axis Resection and Reconstruction for Locally Advanced Pancreatic Cancer. Cancers (Basel) 2024; 16:4115. [PMID: 39682301 DOI: 10.3390/cancers16234115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND With the advent of effective chemotherapy, conversion surgery (CS) has been performed in patients who have responded to pretreatment, even for pancreatic cancer diagnosed as unresectable (UR) at the time of initial diagnosis. In CS, major arterial resection and reconstruction are necessary for complete radical resection. METHODS We discuss the key points for safely performing pancreatectomy with celiac axis (CA) resection combined with reconstruction, divided into resection and arterial reconstruction. The possibility of safe pancreatectomy concurrent with CA resection and reconstruction depends on the ability to create a "golden view" that provides an unimpaired view of the Abdominal Aorta, CA, Superior Mesenteric Artery, Inferior Vena Cava, and left renal vein from the ventral side. Pancreatectomy concurrent with CA resection requires arterial reconstruction. Postoperatively, arterial blood flow must be maintained. To achieve this, tension-free and short bypass should be observed. RESULTS From 2014 to 2024, sixteen URLA patients underwent CS, requiring major artery en bloc resection after pretreatment. We performed DP-CAR in eight patients, gastrectomy-distal pancreatectomy-splenectomy (Appleby procedure) procedure in one patient, PD-CHAR in two patients, PD-CAR in two patients, TP-CAR(spleen preserving) in one patient, and TP-CAR+TG in two patients. In total, five patients required surgery with CA reconstruction. Histopathologically, four of the five patients had T4 pancreatic cancer. The R0 surgical rate was 80%. Complication of Clavien-Dindo IIIa or higher was observed in one patient. There were no deaths. CONCLUSIONS Parallel to the determination of pretreatment, surgeons must be prepared to safely and reliably perform pancreatectomies that require concurrent major arterial resection and reconstruction.
Collapse
Affiliation(s)
- Satoshi Mizutani
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Nobuhiko Taniai
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Makoto Sukegawa
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Takahiro Haruna
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Hiroyasu Furuki
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Hideyuki Takata
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Junji Ueda
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Masato Yoshioka
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Takayuki Aimoto
- Digestive Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Shunichiro Sakamoto
- Department of Cardiovascular Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Kenji Suzuki
- Department of Cardiovascular Surgery, Nippon Medical School Musashikosugi Hospital, 1-383 Kosugimachi, Nakahara, Kawasaki 211-8533, Kanagawa, Japan
| | - Yoshiharu Nakamura
- Department of Surgery, Nippon Medical School Chiba Hokusoh Hospital, 1715 Kamakari, Inzai 270-1694, Chiba, Japan
| | - Hiroshi Yoshida
- Department of Surgery, Nippon Medical School, 1-1-5 Sendagi, Bunkyo, Tokyo 113-8603, Japan
| |
Collapse
|
176
|
Qin D, Xi P, Huang K, Jiang L, Yao Z, Wei R, Li S. Nomogram for predicting post-progression-free survival in patients with recurrent pancreatic ductal adenocarcinoma after radical surgery: a retrospective analysis. Front Med (Lausanne) 2024; 11:1486750. [PMID: 39712186 PMCID: PMC11659012 DOI: 10.3389/fmed.2024.1486750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/20/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Radical resection is the only curative method for patients with pancreatic adenocarcinoma (PDAC). However, nearly 85% of PDAC patients suffer from local or distant recurrence within 5 years after curative resection. The progression of recurrent lesions accelerates the mortality rate in PDAC patients. However, the influence of clinicopathological factors on post-progression-free survival (PPFS), defined as the period from tumor recurrence to the timing of the progression of recurrent lesions, has rarely been discussed. The present study aimed to explore the independent prognostic factors for PPFS and construct a nomogram for PPFS prediction. MATERIALS AND METHODS The 200 recurrent PDAC patients were divided into training and validation groups by leave-one-out cross-validation. The patients' clinicopathological characteristics were compared through a chi-square test. Meanwhile, these factors were enrolled in the univariate and multivariate COX regression to find the independent prognostic factors of PPFS. Moreover, the Kaplan-Meier survival analysis based on the independent prognostic factors was performed. Finally, we constructed a nomogram model for PPFS prediction, followed by an effectiveness examination. RESULTS PDAC patients who received multi-agent chemotherapy after surgery showed a longer PPFS than the single-agent chemotherapy group. PDAC patients who received multi-agent chemotherapy after recurrence showed a similar PPFS compared to the single-agent chemotherapy group. Local recurrence with distant metastases, early recurrence, lympho-vascular invasion, higher T stage, and higher N stage predicted shorter PPFS in recurrent PDAC patients. Finally, a nomogram to indicate the progression of recurrent lesions was constructed. CONCLUSION Multi-agent chemotherapy is recommended for PDAC patients after surgery. Meanwhile, single-agent chemotherapy also deserves consideration after tumor recurrence. Moreover, the nomogram could be used in PPFS prediction.
Collapse
Affiliation(s)
| | | | | | | | | | - Ran Wei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shengping Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
177
|
Zheng H, Yang X, Huang N, Yuan S, Li J, Liu X, Jiang Q, Wu S, Ju Y, Kleeff J, Yin X, Liao Q, Liu Q, Zhao Y. Chimeric antigen receptor macrophages targeting c-MET(CAR-M-c-MET) inhibit pancreatic cancer progression and improve cytotoxic chemotherapeutic efficacy. Mol Cancer 2024; 23:270. [PMID: 39643883 PMCID: PMC11622543 DOI: 10.1186/s12943-024-02184-8] [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: 05/02/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant tumors. Macrophages are abundant in the tumor microenvironment, making them an attractive target for therapeutic intervention. While current immunotherapies, including immune checkpoint inhibition (ICI) and chimeric antigen receptor T (CAR-T) cells, have shown limited efficacy in pancreatic cancer, a novel approach involving chimeric antigen receptor macrophages (CAR-M) has, although promising, not been explored in pancreatic cancer. In this study, we first investigated the role of CAR-M cells targeting c-MET in pancreatic cancer. METHODS The effectiveness and rationality of c-MET as a target for CAR-M in pancreatic cancer were validated through bioinformatic analyses and immunohistochemical staining of samples from pancreatic cancer patients. We utilized flow cytometry and bioluminescence detection methods to demonstrate the specific binding and phagocytic killing effect of CAR-M on pancreatic cancer cells. Additionally, we observed the process of CAR-M engulfing pancreatic cancer cells using confocal microscopy and a long-term fluorescence live cell imaging system. In an in situ tumor model transplanted into NOD/SCID mice, we administered intraperitoneal injections of CAR-M to confirm its inhibitory function on pancreatic cancer. Furthermore, we validated these findings in human monocyte-derived macrophages (hMDM). RESULTS Bioinformatics and tumor tissue microarray analyses revealed significantly higher expression levels of c-MET in tumor tissues, compared to the paired peritumoral tissues, and higher c-MET expression correlated with worse patient survival. CAR-M cells were engineered using human monocytic THP-1 cell line and hMDM targeting c-MET (CAR-M-c-MET). The CAR-M-c-MET cells demonstrated highly specific binding to pancreatic cancer cells and exhibited more phagocytosis and killing abilities than the pro-inflammatory polarized control macrophages. In addition, CAR-M-c-MET cells synergized with various cytotoxic chemotherapeutic drugs. In a NOD/SCID murine model, intraperitoneally injected CAR-M-c-MET cells rapidly migrated to tumor tissue and substantially inhibited tumor growth, which did not lead to obvious side effects. Cytokine arrays and mRNA sequencing showed that CAR-M-c-MET produced higher levels of immune activators than control macrophages. CONCLUSIONS This study provides compelling evidence for the safety and efficacy of CAR-M therapy in treating pancreatic cancer. The results demonstrate that CAR-M-c-MET significantly suppresses pancreatic cancer progression and enhances the effectiveness of cytotoxic chemotherapy. Remarkably, no discernible side effects occur. Further clinical trials are warranted in human pancreatic cancer patients.
Collapse
Affiliation(s)
- Huaijin Zheng
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China
| | - Xinzhe Yang
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Nan Huang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China
| | - Shangqin Yuan
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China
| | - Jiayi Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China
| | - Xudong Liu
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China
| | - Qing Jiang
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Shanshan Wu
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Yue Ju
- Roc Rock Biotechnology (Shenzhen), Shenzhen, 518118, China
| | - Jorg Kleeff
- Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-University Halle-Wittenberg, 06120, Halle (Saale), Germany
| | - Xiushan Yin
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China.
- Roc Rock Biotechnology (Shenzhen), Shenzhen, 518118, China.
| | - Quan Liao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China.
| | - Qiaofei Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Dongcheng District, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuai Fu Yuan, Beijing, 100730, China.
| |
Collapse
|
178
|
Haggstrom L, Chan WY, Nagrial A, Chantrill LA, Sim HW, Yip D, Chin V. Chemotherapy and radiotherapy for advanced pancreatic cancer. Cochrane Database Syst Rev 2024; 12:CD011044. [PMID: 39635901 PMCID: PMC11619003 DOI: 10.1002/14651858.cd011044.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is a lethal disease with few effective treatment options. Many anti-cancer therapies have been tested in the locally advanced and metastatic setting, with mixed results. This review synthesises all the randomised data available to help better inform patient and clinician decision-making. It updates the previous version of the review, published in 2018. OBJECTIVES To assess the effects of chemotherapy, radiotherapy, or both on overall survival, severe or life-threatening adverse events, and quality of life in people undergoing first-line treatment of advanced pancreatic cancer. SEARCH METHODS We searched for published and unpublished studies in CENTRAL, MEDLINE, Embase, and CANCERLIT, and handsearched various sources for additional studies. The latest search dates were in March and July 2023. SELECTION CRITERIA We included randomised controlled trials comparing chemotherapy, radiotherapy, or both with another intervention or best supportive care. Participants were required to have locally advanced, unresectable pancreatic cancer or metastatic pancreatic cancer not amenable to curative intent treatment. Histological confirmation was required. Trials were required to report overall survival. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included 75 studies in the review and 51 in the meta-analysis (11,333 participants). We divided the studies into seven categories: any anti-cancer treatment versus best supportive care; various chemotherapy types versus gemcitabine; gemcitabine-based combinations versus gemcitabine alone; various chemotherapy combinations versus gemcitabine plus nab-paclitaxel; fluoropyrimidine-based studies; miscellaneous studies; and radiotherapy studies. In general, the included studies were at low risk for random sequence generation, detection bias, attrition bias, and reporting bias, at unclear risk for allocation concealment, and high risk for performance bias. Compared to best supportive care, chemotherapy likely results in little to no difference in overall survival (OS) (hazard ratio (HR) 1.08, 95% confidence interval (CI) 0.88 to 1.33; absolute risk of death at 12 months of 971 per 1000 versus 962 per 1000; 4 studies, 298 participants; moderate-certainty evidence). The adverse effects of chemotherapy and impacts on quality of life (QoL) were uncertain. Many of the chemotherapy regimens were outdated. Eight studies compared non-gemcitabine-based chemotherapy regimens to gemcitabine. These showed that 5-fluorouracil (5FU) likely reduces OS (HR 1.69, 95% CI 1.26 to 2.27; risk of death at 12 months of 914 per 1000 versus 767 per 1000; 1 study, 126 participants; moderate certainty), and grade 3/4 adverse events (QoL not reported). Fixed dose rate gemcitabine likely improves OS (HR 0.79, 95% CI 0.66 to 0.94; risk of death at 12 months of 683 per 1000 versus 767 per 1000; 2 studies, 644 participants; moderate certainty), and likely increase grade 3/4 adverse events (QoL not reported). FOLFIRINOX improves OS (HR 0.51, 95% CI 0.43 to 0.60; risk of death at 12 months of 524 per 1000 versus 767 per 1000; P < 0.001; 2 studies, 652 participants; high certainty), and delays deterioration in QoL, but increases grade 3/4 adverse events. Twenty-eight studies compared gemcitabine-based combinations to gemcitabine. Gemcitabine plus platinum may result in little to no difference in OS (HR 0.94, 95% CI 0.81 to 1.08; risk of death at 12 months of 745 per 1000 versus 767 per 1000; 6 studies, 1140 participants; low certainty), may increase grade 3/4 adverse events, and likely worsens QoL. Gemcitabine plus fluoropyrimidine improves OS (HR 0.88, 95% CI 0.81 to 0.95; risk of death at 12 months of 722 per 1000 versus 767 per 1000; 10 studies, 2718 participants; high certainty), likely increases grade 3/4 adverse events, and likely improves QoL. Gemcitabine plus topoisomerase inhibitors result in little to no difference in OS (HR 1.01, 95% CI 0.87 to 1.16; risk of death at 12 months of 770 per 1000 versus 767 per 1000; 3 studies, 839 participants; high certainty), likely increases grade 3/4 adverse events, and likely does not alter QoL. Gemcitabine plus taxane result in a large improvement in OS (HR 0.71, 95% CI 0.62 to 0.81; risk of death at 12 months of 644 per 1000 versus 767 per 1000; 2 studies, 986 participants; high certainty), and likely increases grade 3/4 adverse events and improves QoL. Nine studies compared chemotherapy combinations to gemcitabine plus nab-paclitaxel. Fluoropyrimidine-based combination regimens improve OS (HR 0.79, 95% CI 0.70 to 0.89; risk of death at 12 months of 542 per 1000 versus 628 per 1000; 6 studies, 1285 participants; high certainty). The treatment arms had distinct toxicity profiles, and there was little to no difference in QoL. Alternative schedules of gemcitabine plus nab-paclitaxel likely result in little to no difference in OS (HR 1.10, 95% CI 0.82 to 1.47; risk of death at 12 months of 663 per 1000 versus 628 per 1000; 2 studies, 367 participants; moderate certainty) or QoL, but may increase grade 3/4 adverse events. Four studies compared fluoropyrimidine-based combinations to fluoropyrimidines alone, with poor quality evidence. Fluoropyrimidine-based combinations are likely to result in little to no impact on OS (HR 0.84, 95% CI 0.61 to 1.15; risk of death at 12 months of 765 per 1000 versus 704 per 1000; P = 0.27; 4 studies, 491 participants; moderate certainty) versus fluoropyrimidines alone. The evidence suggests that there was little to no difference in grade 3/4 adverse events or QoL between the two groups. We included only one radiotherapy (iodine-125 brachytherapy) study with 165 participants. The evidence is very uncertain about the effect of radiotherapy on outcomes. AUTHORS' CONCLUSIONS Combination chemotherapy remains standard of care for metastatic pancreatic cancer. Both FOLFIRINOX and gemcitabine plus a taxane improve OS compared to gemcitabine alone. Furthermore, the evidence suggests that fluoropyrimidine-based combination chemotherapy regimens improve OS compared to gemcitabine plus nab-paclitaxel. The effects of radiotherapy were uncertain as only one low-quality trial was included. Selection of the most appropriate chemotherapy for individuals still remains unpersonalised, with clinicopathological stratification remaining elusive. Biomarker development is essential to assist in rationalising treatment selection for patients.
Collapse
Affiliation(s)
- Lucy Haggstrom
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Wei Yen Chan
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Sydney, Australia
| | - Adnan Nagrial
- The Crown Princess Mary Cancer Centre, Westmead, Australia
- Medical School, The University of Sydney, Sydney, Australia
| | - Lorraine A Chantrill
- Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, Australia
- University of Wollongong, Wollongong, Australia
| | - Hao-Wen Sim
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Desmond Yip
- Department of Medical Oncology, The Canberra Hospital, Garran, Australia
- ANU Medical School, Australian National University, Acton, Australia
| | - Venessa Chin
- Medical Oncology, The Kinghorn Cancer Care Centre, St Vincent's Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- Medical Oncology, Garvan Institute of Medical Research, Sydney, Australia
| |
Collapse
|
179
|
Wang D, Cao J, Chen Y, Zhang L, Zhou C, Huang L, Chen Y. Radioresistance-related gene signatures identified by transcriptomics characterize the prognosis and immune landscape of pancreatic cancer patients. BMC Cancer 2024; 24:1497. [PMID: 39639217 PMCID: PMC11619475 DOI: 10.1186/s12885-024-13231-4] [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: 07/31/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Radiotherapy (RT) is an important means of local treatment of solid tumors, and radioresistance is the main reason for RT failure for tumors, especially pancreatic cancer (PC). It is urgent to distinguish key genes and mechanisms of radioresistance in PC. METHODS We acquired the data from The Cancer Genome Atlas (TCGA), obtained the gene modules associated with radioresistance by weighted gene coexpression network analysis (WGCNA), and identified differentially expressed genes (DEGs) between normal and tumor samples. Radioresistance-related genes (RRRGs) were determined with the intersection of WGCNA and DEGs. The hub RRRGs associated with prognosis were distinguished by the least absolute shrinkage and selection operator (LASSO) regression. We established a risk score model using multivariate Cox regression. Immune cell infiltration and drug sensitivity were evaluated through the CIBERSORT algorithm and the "OncoPredict" software package, respectively. The association of the key gene RIC3 and PC clinical features was verified in public databases, and its biological behaviors were explored in vitro. RESULTS The intersection of DEGs and WGCNA confirmed 14 RRRGs, then six hub RRRGs were identified using LASSO. A key four genes (DUSP4, ADORA2B, SCGB2A1, and RIC3) risk score model was constructed and proved to be capable of independently estimating the prognosis of PC. There is no significant difference between risk score groups in various immune cell infiltration and response to immunotherapy. Although the low-risk group seemed to exhibit greater sensitivity to antitumor drugs, the four drugs (5-fluorouracil [5-FU], leucovorin, irinotecan, and oxaliplatin) currently used for PC patients had no statistical difference for the low- and high- group. The overexpression of RIC3 had a synergy effect with irradiation on inhibited malignant biological properties of PC cells, which was verified by detecting the proliferation ability, apoptosis rate, cell cycle distribution, and migration ability of PANC-1 cells. CONCLUSIONS We herein presented signature genes correlated with radioresistance in PC and established a risk score model competent in estimating patients' clinical outcomes and response to antitumor drugs. The above evidence could contribute to comprehending the mechanisms of radioresistance and identifying the underlying therapy targeting.
Collapse
Affiliation(s)
- Dandan Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Jun Cao
- Department of Outpatient, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yanhui Chen
- Department of Neuroscience and Endocrinology, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Lisha Zhang
- Department of Obstetrics, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Chan Zhou
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Litao Huang
- Department of Clinical Research Management, West China Hospital of Sichuan University, Chengdu, China
| | - Yanliang Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
- The First School of Clinical Medicine, Lanzhou University, No. 1, Donggang West Road, Chengguan District, Lanzhou, 730000, China.
| |
Collapse
|
180
|
Robinson RM, Reyes L, Christopher BN, Duncan RM, Burge RA, Siegel J, Nasarre P, Wang P, O'Bryan JP, Hobbs GA, Klauber-DeMore N, Dolloff NG. A High-Affinity Monoclonal Antibody Against the Pancreatic Ductal Adenocarcinoma Target, Anterior Gradient-2 (AGR2/PDIA17). Antibodies (Basel) 2024; 13:101. [PMID: 39727484 DOI: 10.3390/antib13040101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Anterior Gradient-2 (AGR2/PDIA17) is a member of the protein disulfide isomerase (PDI) family of oxidoreductases. AGR2 is up-regulated in several solid tumors, including pancreatic ductal adenocarcinoma (PDAC). Given the dire need for new therapeutic options for PDAC patients, we investigated the expression and function of AGR2 in PDAC and developed a novel series of affinity-matured AGR2-specific single-chain variable fragments (scFvs) and monoclonal antibodies. RESULTS We found that AGR2 was expressed in approximately 90% of PDAC but not normal pancreas biopsies, and the level of AGR2 expression correlated with increasing disease stage. AGR2 expression was inversely related to SMAD4 status in PDAC and colorectal cancer cell models and was secreted from cells into their media. In normal tissues, a high density of AGR2 was detected in the epithelium of cells in the digestive tract but was lacking in most other normal tissue systems. The addition of recombinant AGR2 to cell culture and genetic overexpression of AGR2 increased the adhesion, motility, and invasiveness of both human and mouse PDAC cells. Human phage display library screening led to the discovery of multiple AGR2-specific scFv clones that were affinity-matured to produce monoclonal antibody (MAb) clones with low picomolar binding affinity (S31R/A53F/Y). These high-affinity MAbs inhibited AGR2-mediated cell adhesion, migration, and binding to LYPD3, which is a putative cell surface binding partner of AGR2. CONCLUSIONS Our study provides novel, high-affinity, fully human, anti-AGR2 MAbs that neutralize the pro-tumor effects of extracellular AGR2 in PDAC.
Collapse
Affiliation(s)
- Reeder M Robinson
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Leticia Reyes
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Benjamin N Christopher
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ravyn M Duncan
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Rachel A Burge
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Julie Siegel
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Patrick Nasarre
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - John P O'Bryan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - G Aaron Hobbs
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nancy Klauber-DeMore
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
- MUSC Hollings Cancer Center, Charleston, SC 29425, USA
| | - Nathan G Dolloff
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
- MUSC Hollings Cancer Center, Charleston, SC 29425, USA
- Zucker Institute for Innovation Commercialization, Charleston, SC 29425, USA
| |
Collapse
|
181
|
Roncato R, Bignucolo A, Peruzzi E, Montico M, De Mattia E, Foltran L, Guardascione M, D’Andrea M, Favaretto A, Puglisi F, Swen JJ, Guchelaar HJ, Toffoli G, Cecchin E. Clinical Benefits and Utility of Pretherapeutic DPYD and UGT1A1 Testing in Gastrointestinal Cancer: A Secondary Analysis of the PREPARE Randomized Clinical Trial. JAMA Netw Open 2024; 7:e2449441. [PMID: 39641926 PMCID: PMC11624585 DOI: 10.1001/jamanetworkopen.2024.49441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/14/2024] [Indexed: 12/07/2024] Open
Abstract
Importance To date, the clinical benefit and utility of implementing a DPYD/UGT1A1 pharmacogenetic-informed therapy with fluoropyrimidines and/or irinotecan have not been prospectively investigated. Objective To examine clinically relevant toxic effects, hospitalizations, and related costs while preserving treatment intensity and efficacy outcomes in patients with gastrointestinal cancer. Design, Setting, and Participants This nonprespecified secondary analysis stems from Pre-Emptive Pharmacogenomic Testing for Preventing Adverse Drug Reactions (PREPARE), a multicenter, controlled, open, block-randomized, crossover implementation trial conducted from March 7, 2017, to June 30, 2020, and includes data from Italy according to a sequential study design. The study population included 563 patients (intervention, 252; control [standard of care], 311) with gastrointestinal cancer (age ≥18 years) who were eligible for fluoropyrimidine and/or irinotecan treatment. Data analysis for the present study was performed from May 27 to October 10, 2024. Interventions Participants with actionable variants (DPYD*2A, DPYD*13, .DPYD c.2846A>T, and DPYD c.1236G>A for fluoropyrimidines, and UGT1A1*28, UGT1A1*6, and UGT1A1*27 for irinotecan) received drug or dose adjustments based on Dutch Pharmacogenetics Working Group recommendations. Main Outcomes and Measures The primary outcome was clinically relevant toxic effects (National Cancer Institute Common Terminology Criteria for Adverse Events grade ≥4 hematologic, grade ≥3 nonhematologic, or causing hospitalization, fluoropyrimidines and/or irinotecan causally related). Secondary outcomes included hospitalization rates, toxic effect management costs, intensity of treatment, quality-adjusted life-years, and 3-year overall survival. Results Overall, 1232 patients were enrolled in Italy, with 563 included in this analysis (317 [56.3%] men; median age, 68.0 [IQR, 60.0-75.0] years). In the intervention arm, carriers of any actionable genotype exhibited a 90% lower risk of clinically relevant toxic effects compared with the control arm (odds ratio, 0.1; 95% CI, 0.0-0.8; P = .04). They also presented higher toxic effect management costs per patient ($4159; 95% CI, $1510-$6810) compared with patients in the intervention arm ($26; 95% CI, 0-$312) (P = .004) and a higher rate of hospitalization (34.8% vs 11.8%; P = .12). The differences were not significant among all patients. Three-year overall survival did not differ significantly between arms, while quality-adjusted life-years significantly improved in the intervention arm. The pharmacogenetics-informed approach did not manifest a detrimental effect on treatment intensity in actionable genotype carriers. Conclusions and Relevance In this secondary analysis of PREPARE, pretreatment application of DPYD- and UGT1A1-guided treatment appeared to increase safety and reduce hospitalizations and related costs in patients with gastrointestinal cancer. Clinical benefit did not appear to be affected. Trial Registration ClinicalTrials.gov Identifier: NCT03093818.
Collapse
Affiliation(s)
- Rossana Roncato
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
- Department of Medicine, University of Udine, Udine, Italy
| | - Alessia Bignucolo
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Elena Peruzzi
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Marcella Montico
- Clinical Trial Office, Scientific Direction, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Luisa Foltran
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Michela Guardascione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Mario D’Andrea
- Department of Medical Oncology, Ospedale San Paolo / Ospedale Padre Pio, Civitavecchia, Rome, Italy
| | - Adolfo Favaretto
- Department of Medical Oncology, Azienda ULSS 2 Marca Trevigiana Distretto di Treviso, Treviso, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Udine, Italy
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Jesse Joachim Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| |
Collapse
|
182
|
Yamaguchi N, Wu YG, Ravetch E, Takahashi M, Khan AG, Hayashi A, Mei W, Hsu D, Umeda S, de Stanchina E, Lorenz IC, Iacobuzio-Donahue CA, Tavazoie SF. A Targetable Secreted Neural Protein Drives Pancreatic Cancer Metastatic Colonization and HIF1α Nuclear Retention. Cancer Discov 2024; 14:2489-2508. [PMID: 39028915 PMCID: PMC11611693 DOI: 10.1158/2159-8290.cd-23-1323] [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: 11/13/2023] [Revised: 05/29/2024] [Accepted: 07/18/2024] [Indexed: 07/21/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an increasingly diagnosed cancer that kills 90% of afflicted patients, with most patients receiving palliative chemotherapy. We identified neuronal pentraxin 1 (NPTX1) as a cancer-secreted protein that becomes overexpressed in human and murine PDAC cells during metastatic progression and identified adhesion molecule with Ig-like domain 2 (AMIGO2) as its receptor. Molecular, genetic, biochemical, and pharmacologic experiments revealed that secreted NPTX1 acts cell-autonomously on the AMIGO2 receptor to drive PDAC metastatic colonization of the liver-the primary site of PDAC metastasis. NPTX1-AMIGO2 signaling enhanced hypoxic growth and was critically required for hypoxia-inducible factor-1α (HIF1α) nuclear retention and function. NPTX1 is overexpressed in human PDAC tumors and upregulated in liver metastases. Therapeutic targeting of NPTX1 with a high-affinity monoclonal antibody substantially reduced PDAC liver metastatic colonization. We thus identify NPTX1-AMIGO2 as druggable critical upstream regulators of the HIF1α hypoxic response in PDAC. Significance: We identified the NPTX1-AMIGO2 axis as a regulatory mechanism upstream of HIF1α-driven hypoxia response that promotes PDAC liver metastasis. Therapeutic NPTX1 targeting outperformed a common chemotherapy regimen in inhibiting liver metastasis and suppressed primary tumor growth in preclinical models, revealing a novel therapeutic strategy targeting hypoxic response in PDAC.
Collapse
Affiliation(s)
- Norihiro Yamaguchi
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Y Gloria Wu
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Ethan Ravetch
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Mai Takahashi
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Abdul G. Khan
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
| | - Akimasa Hayashi
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wenbin Mei
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Dennis Hsu
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Shigeaki Umeda
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Ivo C. Lorenz
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
| | | | - Sohail F. Tavazoie
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
183
|
Mouawad A, Habib S, Boutros M, Attieh F, Kourie HR. How to find a needle in a haystack: a systematic review on targeting KRAS wild-type pancreatic cancer. Future Oncol 2024; 20:3539-3547. [PMID: 38861291 PMCID: PMC11776852 DOI: 10.1080/14796694.2024.2355078] [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: 01/21/2024] [Accepted: 05/10/2024] [Indexed: 06/12/2024] Open
Abstract
Aim: Pancreatic adenocarcinoma is a very aggressive type of cancer, in which targeted therapies have not yet been fully utilized. KRAS wild-type pancreatic adenocarcinoma tumors are associated with different genomic alterations in comparison to KRAS mutated pancreatic adenocarcinoma. Objective: This systematic review aims to provide a one-stop summary of all these alterations, their proposed targeted treatment and their effect on disease progression. Methods: An electronic search strategy was elaborated in the PubMed database between 2020 and January 2024. Results: 21 studies were included, and we found that the most frequent targetable genomic alterations in KRAS wild-type pancreatic adenocarcinoma were BRAF, EGFR, FGFR, MSI-H/dMMR, Her2/ERBB2 amplification, BRCA1/2 and other HRDs, and gene fusions like ALK, NTRK and NRG1.
Collapse
Affiliation(s)
- Antoine Mouawad
- Université Saint-Joseph de Beyrouth, Beyrouth, 11-5076, Lebanon
| | - Sofia Habib
- Université Saint-Joseph de Beyrouth, Beyrouth, 11-5076, Lebanon
| | - Marc Boutros
- Université Saint-Joseph de Beyrouth, Beyrouth, 11-5076, Lebanon
| | - Fouad Attieh
- Université Saint-Joseph de Beyrouth, Beyrouth, 11-5076, Lebanon
| | - Hampig Raphaël Kourie
- Department of Hematology-Oncology, Université Saint-Joseph de Beyrouth, Beyrouth, 11-5076, Lebanon
| |
Collapse
|
184
|
Vošmik M, John S, Dvořák J, Mohelníková-Duchoňová B, Melichar B, Lohynská R, Ryška A, Banni AM, Krempová J, Sirák I. Stereotactic Radiotherapy Plus Nivolumab in Patients with Locally Advanced Pancreatic Cancer: Results from Phase 1/2 Clinical CA209-9KH Trial. Oncol Ther 2024; 12:817-831. [PMID: 39441483 PMCID: PMC11574225 DOI: 10.1007/s40487-024-00309-z] [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: 08/07/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION The dismal prognosis of pancreatic ductal adenocarcinoma (PDAC) highlights the urgent need for novel therapeutic strategies. Immune checkpoint inhibitors (ICIs) seem to be ineffective in most PDAC studies. Therefore, we conducted an open-label, multicenter phase 1/2 study (CA209-9KH) to evaluate the safety of stereotactic radiotherapy (SRT) and sequential ICI therapy in PDAC, as well as to validate the efficacy of this regimen as a potential activator of antitumor immunity. METHODS Patients aged ≥ 18 years with unresectable non-metastatic PDAC following four FOLFIRINOX induction cycles were included. Treatment comprised SRT (4 × 8 Gy) and sequential nivolumab administration until disease progression or unacceptable toxicity. The primary endpoints were safety and toxicity assessment. Secondary endpoints included progression-free survival (PFS), overall survival (OS), biomarker evaluation, and quality of life (QoL) analysis. RESULTS Twenty-two patients were screened, with 15 enrolled. Eleven (median) nivolumab cycles were administered. SRT demonstrated low and clinically nonsignificant toxicity, whereas nivolumab toxicity aligned with prior safety profiles, without grade 4-5 events observed. Three patients discontinued therapy owing to toxicity. Median PFS and OS were 8.1 and 13.0 months, respectively, with 12-month PFS and OS rates of 0% and 66.7%, respectively, and a 24-month OS rate of 8.9%. Biomarker levels correlated with clinical or radiological disease control. Patient-reported QoL remained acceptable, deteriorating with disease progression. CONCLUSION SRT and nivolumab therapy exhibited manageable toxicity profiles consistent with previous findings; however, long-term treatment responses were not achieved with this regimen in locally advanced PDAC. Another strategy to trigger antitumor immunity in PDAC needs to be sought. TRIAL REGISTRATION EudraCT: 2017-003404-52; ClinicalTrials.gov: NCT04098432.
Collapse
Affiliation(s)
- Milan Vošmik
- Department of Oncology and Radiotherapy, University Hospital Hradec Králové, Sokolská 581, 50005, Hradec Králové, Czech Republic.
- Faculty of Medicine in Hradec Králové, Charles University, Prague, Czech Republic.
| | - Stanislav John
- Department of Oncology and Radiotherapy, University Hospital Hradec Králové, Sokolská 581, 50005, Hradec Králové, Czech Republic
- Faculty of Medicine in Hradec Králové, Charles University, Prague, Czech Republic
| | - Josef Dvořák
- Department of Oncology, Thomayer University Hospital, Prague, Czech Republic
- First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Beatrice Mohelníková-Duchoňová
- Department of Oncology, University Hospital Olomouc, Olomouc, Czech Republic
- Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, University Hospital Olomouc, Olomouc, Czech Republic
- Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Radka Lohynská
- Department of Oncology, Thomayer University Hospital, Prague, Czech Republic
- First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Aleš Ryška
- Faculty of Medicine in Hradec Králové, Charles University, Prague, Czech Republic
- The Fingerland Department of Pathology, University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Aml Mustafa Banni
- Department of Oncology and Radiotherapy, University Hospital Hradec Králové, Sokolská 581, 50005, Hradec Králové, Czech Republic
- Faculty of Medicine in Hradec Králové, Charles University, Prague, Czech Republic
| | - Johana Krempová
- Department of Oncology and Radiotherapy, University Hospital Hradec Králové, Sokolská 581, 50005, Hradec Králové, Czech Republic
| | - Igor Sirák
- Department of Oncology and Radiotherapy, University Hospital Hradec Králové, Sokolská 581, 50005, Hradec Králové, Czech Republic
- Faculty of Medicine in Hradec Králové, Charles University, Prague, Czech Republic
| |
Collapse
|
185
|
Tang H, Zhu Z, Ying J, You T, Ge H, Cheng Y, Geng R, Zhou N, Qiu W, Ge Y, Wang Y, Bai C. The prognostic role of WeChat-based instant doctor-patient communication in patients with advanced pancreatic cancer. J Cancer Surviv 2024; 18:1903-1911. [PMID: 37460857 DOI: 10.1007/s11764-023-01429-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/05/2023] [Indexed: 10/25/2024]
Abstract
BACKGROUND Patients with advanced pancreatic ductal adenocarcinoma (PDAC) need timely medical assistance since the emergence of complications due to the disease and antitumor treatment. Studies have confirmed that instant messaging can improve the quality of life and compliance of cancer patients. However, the prognostic role of instant doctor-patient communication based on instant messaging applications in PDAC has not been studied. METHODS Patients with PDAC who received first-line chemotherapy at Peking Union Medical College Hospital between January 2015 and October 2022 were reviewed. We categorized patients into two groups according to whether they received WeChat-based instant doctor-patient communication, and the prognosis and toxicity data between the two groups were compared. RESULTS A total of 431 PDAC patients were enrolled, of whom 163 had long-term instant communication with their doctors based on WeChat, and 268 did not receive WeChat-based instant communication. There was no significant correlation between WeChat-based communication and first-line chemotherapy overall response rate (14.1% vs. 8.6%, p = 0.074), incidence of grade ≥ 3 adverse events (66.9% vs. 65.7%, p = 0.814) or overall survival (14.7 vs. 13.9 months, p = 0.170) in all enrolled patients. However, patients who received WeChat-based instant communication had a higher completion rate of first-line chemotherapy (42.0% vs. 30.7%, p = 0.023). Consistently, in the patients who developed grade ≥ 3 adverse events (n = 231), those who received WeChat-based instant communication had significantly longer overall survival (17.3 vs. 15.3 months, p = 0.018), even after adjustment for biases. CONCLUSIONS WeChat-based instant doctor-patient communication demonstrated no superiority in improving the efficacy of chemotherapy or preventing chemotherapy toxicity in PDAC patients, but it may contribute to improving the completion rate of chemotherapy and the prognosis in patients who experienced severe adverse events. IMPLICATIONS FOR CANCER SURVIVORS Instant doctor‒patient communication may help to timely and appropriately deal with adverse events and prolong the survival time of patients with PDAC, supporting the promotion of mobile technology in clinical practice.
Collapse
Affiliation(s)
- Hui Tang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhou Zhu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jinrong Ying
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tingting You
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hui Ge
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ruixuan Geng
- Department of International Medical Services, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Na Zhou
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Qiu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuping Ge
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yingyi Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
186
|
Morimoto Y, Takada K, Nakano A, Takeuchi O, Watanabe K, Hirohara M, Masuda Y. Combination of S-1 and the oral ATR inhibitor ceralasertib is effective against pancreatic cancer cells. Cancer Chemother Pharmacol 2024; 94:763-774. [PMID: 39271497 DOI: 10.1007/s00280-024-04716-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
PURPOSE In our previous study, we found that the Chk1 inhibitor prexasertib enhances the antitumour effect of the oral anticancer drug S-1 against pancreatic cancer cells. In this study, we investigated the effect of combining S-1 and ceralasertib, an oral inhibitor of ATR, which is located upstream of Chk1. Ceralasertib is currently being investigated in multiple clinical trials for various cancers. METHODS The cell-proliferation inhibitory effect was measured by MTT assay, using the pancreatic cancer cell lines BxPC-3, SUIT-2, PANC-1, and MIA PaCa-2, while apoptosis was measured by flow cytometry using PI/Annexin staining. The mechanism underlying the combined effect was analysed using western blotting, and the antitumor effect was analysed using a mouse xenograft model. RESULTS MTT assay revealed that the combination of S-1 and ceralasertib had a synergistic effect, leading to the suppression of cell proliferation. Measurement with PI/Annexin staining revealed that the combination of S-1 and ceralasertib induced apoptosis more efficiently than either drug alone. Western blotting results showed that ceralasertib inhibited S-1-induced activation of ATR and Chk1. The average estimated tumour volume after 3 weeks of administration was 601 mm3 in the S-1 group, 580 mm3 in the ceralasertib group, and 298 mm3 in the combination group. CONCLUSION The combination of S-1 and ceralasertib demonstrated a high antiproliferative effect in inhibiting tumour growth in vitro.
Collapse
Affiliation(s)
- Yoshihito Morimoto
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan.
| | - Kimihiko Takada
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Ami Nakano
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Osamu Takeuchi
- BioMedical Laboratory, Department of Research, Kitasato Institute Hospital, Tokyo, 108-8642, Japan
| | - Kazuhiro Watanabe
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Masayoshi Hirohara
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Yutaka Masuda
- Center for Education and Research on Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| |
Collapse
|
187
|
Gu J, Shi Z, Zhou S, Zhou Q, Nie S, Li H, Shen S, Zou X. Triptolide exhibits dual anti-tumor effects through inhibiting autophagy and extracellular matrix activation in pancreatic cancer. J Cancer Res Ther 2024; 20:2041-2054. [PMID: 39792414 DOI: 10.4103/jcrt.jcrt_186_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 09/10/2024] [Indexed: 01/12/2025]
Abstract
AIM The tumor microenvironment in pancreatic cancer, characterized by abundant desmoplastic stroma, has been implicated in the failure of chemotherapy. Therefore, developing therapeutic strategies targeting tumor and stromal cells is essential. Triptolide, a natural compound derived from the plant Tripterygium wilfordii, has shown antitumor activity in various cancers, including pancreatic cancer. However, its effects on pancreatic cancer cells and the microenvironment remain unclear. This study aimed to explore the effect of triptolide on tumor cells and the tumor microenvironment in pancreatic cancer. METHODS Cell Counting Kit-8, colony formation, apoptosis, and cell cycle assays were performed to determine the effect of triptolide on tumor cells. Additionally, co-culture assays were performed to explore the effects of the compound on cancer-associated fibroblasts (CAFs) in vitro. Orthotopic xenograft and subcutaneous tumor models were used to explore the antitumor and antistromal activation effects of triptolide in vivo. RNA sequencing was performed to identify the pathways involved in these processes in pancreatic cancer cells. RESULTS Triptolide inhibited the proliferation of pancreatic cancer cells and attenuated stromal activation in vitro and in vivo. Furthermore, it suppressed autophagy and induced apoptosis in pancreatic cancer cells by inhibiting the secretion of CXCL1. Extracellular matrix formation in CAFs was disrupted by suppressing the paracrine secretion of TGF-β from tumor cells. CONCLUSION These findings indicate that triptolide plays a dual antitumor role against tumor cells and CAFs, thus providing new insights into treating pancreatic cancer in the future.
Collapse
Affiliation(s)
- Jianxiang Gu
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Gastroenterology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Zhao Shi
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Siqi Zhou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Quan Zhou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Shuang Nie
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Hongzhen Li
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Shanshan Shen
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiaoping Zou
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
- Department of Gastroenterology, Affiliated Taikang Xianlin Drum Tower Hospital, Medical school of Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
188
|
Halle-Smith JM, Leung P, Hall L, Aksin M, van Laarhoven S, Skipworth J, Chatzizacharias N, Brown RM, Roberts KJ. Factors associated with favourable pathological tumour response after neoadjuvant chemotherapy in patients with pancreatic ductal adenocarcinoma. HPB (Oxford) 2024; 26:1536-1543. [PMID: 39384505 DOI: 10.1016/j.hpb.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 10/11/2024]
Abstract
INTRODUCTION Pathological response of pancreatic ductal adenocarcinoma (PDAC) to neoadjuvant chemotherapy (NAT) has been associated with oncological outcome. The aim of the study was to investigate factors associated with favourable tumour regression in patients undergoing pancreatic resection for PDAC. METHODS Patients who received NAT before undergoing PDAC resection at two institutions were reviewed. Tumour regression grading (TRG) was scored according to the College of American Pathologists (CAP) system. Interactions between chemotherapy, tumour and surgical factors with TRG were explored. RESULTS 54 patients were identified, with 12 (22%) displaying a favourable response to NAT. The type of chemotherapy agent received, the number of cycles or a dose reduction during NAT course was not significantly different between the groups. The time from diagnosis to chemotherapy and time from end of chemotherapy to surgery were also similar between the groups. A favourable TRG was associated with greater disease-free survival median 33.2 months vs. 10.3 months; p = 0.0) but not overall survival (median 43.8 months vs. 32.3 months; p = 0.200), which may be due to small sample size. CONCLUSIONS Chemotherapy factors were not significantly related to a favourable response to NAT. Future studies should seek to identify modifiable factors associated with a favourable TRG.
Collapse
Affiliation(s)
- James M Halle-Smith
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, West Midlands, United Kingdom; College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, United Kingdom.
| | - Prudence Leung
- Aston University, Birmingham, West Midlands, United Kingdom
| | - Lewis Hall
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, West Midlands, United Kingdom
| | - Merve Aksin
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, West Midlands, United Kingdom
| | - Stijn van Laarhoven
- Hepatobiliary and Pancreatic Surgery Unit, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - James Skipworth
- Hepatobiliary and Pancreatic Surgery Unit, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Nikolaos Chatzizacharias
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, West Midlands, United Kingdom
| | - Rachel M Brown
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, West Midlands, United Kingdom; College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, United Kingdom
| | - Keith J Roberts
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, West Midlands, United Kingdom; College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, United Kingdom
| |
Collapse
|
189
|
Steadman JA, Sultan A, Day CN, Parish MA, Warner SG, Kendrick ML, Truty MJ, Jin Z, Thiels CA. Impact of proton pump inhibitors on pathologic response rates following fluoropyrimidine-based neoadjuvant chemotherapy in pancreatic cancer patients. J Surg Oncol 2024; 130:1634-1642. [PMID: 39257300 DOI: 10.1002/jso.27837] [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: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Proton pump inhibitors (PPIs) negatively impact fluoropyrimidine-based chemotherapy efficacy in colorectal cancer. This study assessed PPI impact on major pathologic response (mPR) rates of pancreatic adenocarcinoma (PDAC) patients receiving fluoropyrimidine-based chemotherapy. METHODS An institutional retrospective review of resected PDAC patients receiving neoadjuvant fluoropyrimidine-based chemotherapy (98% FOLFIRINOX) from 2011 to 2021 was conducted. Outcomes were stratified by use or nonuse of PPIs within 6 months of neoadjuvant chemotherapy initiation. Primary outcome was mPR defined as complete or near complete response. RESULTS Among 540 patients included, the median age was 64 (IQR: 60-70) years, 297 (55%) were male, and 202 (37%) were PPI users. 170 (31%) patients had mPR with similar rates among PPI users and nonusers (29% vs. 33%, p = 0.38). No difference in mPR was seen between PPI users and nonusers receiving chemoradiation (35% vs. 36%, p = 0.89) or ≥8 cycles of NAC (33% vs. 36%, p = 0.55). Median OS for PPI users was 30.9 versus 31.7 months for nonusers (p = 0.62). On multivariable analysis, PPI therapy was not associated with decreased survival. CONCLUSION PPI usage did not significantly influence mPR or OS following neoadjuvant fluoropyrimidine-based chemotherapy in resected PDAC patients. Further analysis of all patients, not just those who underwent resection, is required.
Collapse
Affiliation(s)
- Jessica A Steadman
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ahmer Sultan
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Courtney N Day
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Marie A Parish
- Division of Pharmacy Cancer Care, Mayo Clinic, Rochester, Minnesota, USA
| | - Susanne G Warner
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael L Kendrick
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark J Truty
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Zhaohui Jin
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Cornelius A Thiels
- Division of Hepatobiliary & Pancreas Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
190
|
Eyuboglu S, Alpsoy S, Uversky VN, Coskuner-Weber O. Key genes and pathways in the molecular landscape of pancreatic ductal adenocarcinoma: A bioinformatics and machine learning study. Comput Biol Chem 2024; 113:108268. [PMID: 39467488 DOI: 10.1016/j.compbiolchem.2024.108268] [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: 10/03/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is recognized for its aggressive nature, dismal prognosis, and a notably low five-year survival rate, underscoring the critical need for early detection methods and more effective therapeutic approaches. This research rigorously investigates the molecular mechanisms underlying PDAC, with a focus on the identification of pivotal genes and pathways that may hold therapeutic relevance and prognostic value. Through the construction of a protein-protein interaction (PPI) network and the examination of differentially expressed genes (DEGs), the study uncovers key hub genes such as CDK1, KIF11, and BUB1, demonstrating their substantial role in the pathogenesis of PDAC. Notably, the dysregulation of these genes is consistent across a spectrum of cancers, positing them as potential targets for wide-ranging cancer therapeutics. This study also brings to the fore significant genes encoding intrinsically disordered proteins, in particular GPRC5A and KRT7, unveiling promising new pathways for therapeutic intervention. Advanced machine learning techniques were harnessed to classify PDAC patients with high accuracy, utilizing the key genetic markers as a dataset. The Support Vector Machine (SVM) model leveraged the hub genes to achieve a sensitivity of 91 % and a specificity of 85 %, while the RandomForest model notched a sensitivity of 91 % and specificity of 92.5 %. Crucially, when the identified genes were cross-referenced with TCGA-PAAD clinical datasets, a tangible correlation with patient survival rates was discovered, reinforcing the potential of these genes as prognostic biomarkers and their viability as targets for therapeutic intervention. This study's findings serve as a potent testament to the value of molecular analysis in enhancing the understanding of PDAC and in advancing the pursuit for more effective diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Sinan Eyuboglu
- Turkish-German University, Molecular Biotechnology, Sahinkaya Caddesi, No. 106, Beykoz, Istanbul 34820, Turkey
| | - Semih Alpsoy
- Turkish-German University, Molecular Biotechnology, Sahinkaya Caddesi, No. 106, Beykoz, Istanbul 34820, Turkey
| | - Vladimir N Uversky
- USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Orkid Coskuner-Weber
- Turkish-German University, Molecular Biotechnology, Sahinkaya Caddesi, No. 106, Beykoz, Istanbul 34820, Turkey.
| |
Collapse
|
191
|
Ito R, Yoshioka R, Yanagisawa N, Ishii S, Sugitani J, Furuya R, Fujisawa M, Imamura H, Mise Y, Isayama H, Saiura A. Survival Analysis of Conversion Surgery in Borderline Resectable and Locally Advanced Unresectable Pancreatic Ductal Adenocarcinoma Addressing Selection and Immortal Time Bias: A Retrospective Single-Center Study. Ann Surg Oncol 2024; 31:8744-8755. [PMID: 39361176 DOI: 10.1245/s10434-024-16203-x] [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: 05/23/2024] [Accepted: 09/03/2024] [Indexed: 11/10/2024]
Abstract
BACKGROUND The purpose of this study was to provide a detailed evaluation of the oncological advantages of surgery following neoadjuvant chemotherapy (NAC) for patients with borderline resectable (BR) or unresectable (UR) pancreatic ductal adenocarcinoma (PDAC), with a focus on minimizing biases. Recently, NAC has become the standard care for BR or UR locally advanced (UR-LA) PDAC, however, many studies have assessed survival benefits and favorable variables without consideration for biases, particularly immortal time bias. PATIENTS AND METHODS This study included patients diagnosed with BR or UR-LA PDAC at Juntendo University Hospital from 2019 to 2022. To mitigate bias, we applied methods such as propensity score matching (PSM), time-dependent covariate Cox proportional hazard regression analysis (TDC), landmark analysis, and multivariable Cox proportional hazards regression model. RESULTS The study analyzed 124 patients, dividing them into a surgery group (n = 57) and a chemotherapy-only group (n = 67). After PSM, there were 21 matched pairs. Survival analysis using TDC analysis showed that the surgery group had significantly better overall survival compared with the chemotherapy-only group in both the entire cohort and the matched pairs. Cox regression analysis of the entire cohort also revealed a similar superiority of surgery, while the landmark analysis showed varying results depending on the landmark setting. CONCLUSIONS After careful adjustment for selection and immortal time biases, surgery following NAC appears to significantly extend survival in patients with BR or UR PDAC.
Collapse
Affiliation(s)
- Ryota Ito
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ryuji Yoshioka
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Naotake Yanagisawa
- Medical Technology Innovation Centre, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shigeto Ishii
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jun Sugitani
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ryoji Furuya
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masahiro Fujisawa
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroshi Imamura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshihiro Mise
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroyuki Isayama
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akio Saiura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
192
|
Wainberg ZA, M O'Reilly E. NALIRIFOX in the frontline for metastatic pancreatic cancer: evidence beyond NAPOLI 3. Nat Rev Clin Oncol 2024; 21:900-901. [PMID: 39455872 DOI: 10.1038/s41571-024-00952-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Affiliation(s)
- Zev A Wainberg
- David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Eileen M O'Reilly
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
193
|
Mastrantoni L, Chiaravalli M, Spring A, Beccia V, Di Bello A, Bagalà C, Bensi M, Barone D, Trovato G, Caira G, Giordano G, Bria E, Tortora G, Salvatore L. Comparison of first-line chemotherapy regimens in unresectable locally advanced or metastatic pancreatic cancer: a systematic review and Bayesian network meta-analysis. Lancet Oncol 2024; 25:1655-1665. [PMID: 39542008 DOI: 10.1016/s1470-2045(24)00511-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND In advanced pancreatic ductal adenocarcinoma (PDAC), first-line chemotherapy is the standard of care. Due to the absence of head-to-head comparisons in clinical trials, we performed this systematic review and network meta-analysis to compare treatment options for PDAC in terms of their efficacy and toxicity. METHODS PubMed, the Cochrane Central Register of Controlled Trials, Embase, and oncological meetings websites were searched until Nov 15, 2023. We included phase 2-3 randomised controlled trials published after Jan 1, 2000, evaluating first-line treatments in patients with previously untreated, unresectable, locally advanced or metastatic PDAC. Primary endpoints assessed were progression-free survival and overall survival. Summary data were extracted from published reports. The deviance information criterion was used to choose between a random-effects or fixed-effects model. Hazard ratios (HRs) with 95% credible intervals were estimated using a Bayesian approach. The risk of bias was evaluated using the Cochrane Risk of Bias 2 (RoB 2) tool and studies were graded as low, some concerns, or high risk of bias. The quality of evidence was evaluated using the Grading of Recommendations Assessment, Development, and Evaluation approach. This systematic review and network meta-analysis is registered with PROSPERO, CRD42023450330. FINDINGS 6050 records were screened and 79 randomised controlled trials (22 168 patients) were included in the analysis. Gemcitabine was the most frequent comparator (in 50 [63%] of 79 trials) and was considered as the reference treatment. A fixed-effect model was used to analyse the primary outcomes. Regarding progression-free survival (71 trials, 19 479 patients), the most effective treatments were gemcitabine plus nab-paclitaxel alternating folinic acid, fluorouracil, and oxaliplatin ([FOLFOX] HR 0·32, 95% credible interval 0·22-0·47), cisplatin, nab-paclitaxel, capecitabine, and gemcitabine ([PAXG] 0·35, 0·22-0·55), and liposomal irinotecan in combination with fluorouracil, leucovorin, and oxaliplatin ([NALIRIFOX] 0·43, 0·34-0·54), followed by fluorouracil, leucovorin, irinotecan, and oxaliplatin ([FOLFIRINOX] 0·55, 0·47-0·65) and gemcitabine plus nab-paclitaxel (0·62, 0·54-0·72). Similar results were observed for overall survival (79 trials, 22 104 patients): PAXG (HR 0·40, 95% credible interval 0·25-0·65), gemcitabine plus nab-paclitaxel alternating FOLFOX (0·46, 0·32-0·66), and NALIRIFOX (0·56, 0·45-0·70) had the highest benefit, followed by FOLFIRINOX (0·66, 0·56-0·78) and gemcitabine plus nab-paclitaxel (0·67, 0·59-0·77). The overall risk of bias was low to some concerns. Certainty of evidence was low. INTERPRETATION Our findings suggest that NALIRIFOX and FOLFIRINOX should be the preferred options for patients who can tolerate these regimens, with gemcitabine plus nab-paclitaxel remaining a viable alternative, particularly in patients unfit for triplet therapy. Phase 3 randomised controlled trials investigating concomitant or sequential quadruplets are warranted. FUNDING None.
Collapse
Affiliation(s)
- Luca Mastrantoni
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Marta Chiaravalli
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Alexia Spring
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Viria Beccia
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Armando Di Bello
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Cinzia Bagalà
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Maria Bensi
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Diletta Barone
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Giovanni Trovato
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Giulia Caira
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Giulia Giordano
- Department of Aging, Orthopedics and Rheumatological Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Emilio Bria
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy; Medical Oncology Unit, Ospedale Isola Tiberina-Gemelli Isola, Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy
| | - Lisa Salvatore
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS Rome, Italy.
| |
Collapse
|
194
|
Rémond M, Smolenschi C, Tarabay A, Gelli M, Fernandez-de-Sevilla E, Mouawia A, Cosconea S, Tselikas L, Barbe R, Fuerea A, Bani MA, Deloger M, Besse B, Pudlarz T, Valéry M, Boige V, Hollebecque A, Ducreux M, Boilève A. Clinical and molecular features of early onset pancreatic adenocarcinoma. Int J Cancer 2024; 155:1969-1981. [PMID: 39146492 DOI: 10.1002/ijc.35135] [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: 01/26/2024] [Revised: 04/19/2024] [Accepted: 05/07/2024] [Indexed: 08/17/2024]
Abstract
Pancreatic adenocarcinoma (PDAC) is a major health burden and may become the second cause of death by cancer in developed countries. The incidence of early-onset pancreatic cancer (EOPC, defined by an age at diagnosis <50 years old) is increasing. Here, we conducted a study of all PDAC patients followed at our institution. Patients were classified as EOPC or non-early onset (nEOPC, >50). Eight hundred and seventy eight patients were included, of which 113 EOPC, exhibiting a comparable performance status. EOPC were more often diagnosed at the metastatic stage (70.0% vs 58.3%) and liver metastases were more prevalent at diagnosis (60.2% vs. 43.9%). The median overall survival (OS) from diagnosis was 18.1 months, similar between EOPC and nEOPC. Among patients who underwent surgery, recurrence-free survival was similar between age groups. Among metastatic patients, first line progression free survival was similar but EOPC received more treatment lines (72.3% vs. 58.1% received ≥2 lines). Regarding molecular alterations, the mean tumor mutational burden (TMB) was lower in EOPC (1.42 vs. 2.95 mut/Mb). The prevalence of KRAS and BRCA1/2 mutations was similar, but EOPC displayed fewer alterations in CNKN2A/B. Fifty eight patients (18.6%) had actionable alterations (ESCAT I-III) and 31 of them received molecularly matched treatments. On the transcriptomic level, despite its clinical aggressiveness, EOPC was less likely to display a basal-like phenotype. To conclude, EOPC were diagnosed more frequently at the metastatic stage. OS and 1st line PFS were similar to nEOPC. EOPC displayed specific molecular features, such as a lower TMB and fewer alterations in CDKN2A/B.
Collapse
Affiliation(s)
- Maxime Rémond
- Département de Médecine, Gustave Roussy, Villejuif, France
| | - Cristina Smolenschi
- Département de Médecine, Gustave Roussy, Villejuif, France
- Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy, Villejuif, France
| | | | | | | | - Ali Mouawia
- Département de Médecine, Gustave Roussy, Villejuif, France
| | | | - Lambros Tselikas
- Département de Radiologie Interventionnelle, Gustave Roussy, Villejuif, France
| | - Remy Barbe
- Département d'Imagerie, Gustave Roussy, Villejuif, France
| | - Alina Fuerea
- Département de Médecine, Gustave Roussy, Villejuif, France
| | - Mohamed A Bani
- Département d'Anatomopathologie, Gustave Roussy, Villejuif, France
| | - Marc Deloger
- Service de Bioinformatique, Gustave Roussy, Villejuif, France
| | - Benjamin Besse
- Département de Médecine, Gustave Roussy, Villejuif, France
- Université Paris Saclay, Orsay, France
| | - Thomas Pudlarz
- Département de Médecine, Gustave Roussy, Villejuif, France
| | - Marine Valéry
- Département de Médecine, Gustave Roussy, Villejuif, France
| | - Valérie Boige
- Département de Médecine, Gustave Roussy, Villejuif, France
| | - Antoine Hollebecque
- Département de Médecine, Gustave Roussy, Villejuif, France
- Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy, Villejuif, France
| | - Michel Ducreux
- Département de Médecine, Gustave Roussy, Villejuif, France
- Université Paris Saclay, Orsay, France
| | - Alice Boilève
- Département de Médecine, Gustave Roussy, Villejuif, France
- Université Paris Saclay, Orsay, France
| |
Collapse
|
195
|
Chao T, Wang ZX, Bowne WB, Yudkoff CJ, Torjani A, Swaminathan V, Kavanagh TR, Roadarmel A, Sholevar CJ, Cannaday S, Krampitz G, Zhan T, Gorgov E, Nevler A, Lavu H, Yeo CJ, Peiper SC, Jiang W. Association of Mutant KRAS Alleles With Morphology and Clinical Outcomes in Pancreatic Ductal Adenocarcinoma. Arch Pathol Lab Med 2024; 148:1299-1309. [PMID: 38452805 DOI: 10.5858/arpa.2023-0005-oa] [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] [Accepted: 12/21/2023] [Indexed: 03/09/2024]
Abstract
CONTEXT.— Mutant KRAS is the main oncogenic driver in pancreatic ductal adenocarcinomas (PDACs). However, the clinical and phenotypic implications of harboring different mutant KRAS alleles remain poorly understood. OBJECTIVE.— To characterize the potential morphologic and clinical outcome differences in PDACs harboring distinct mutant KRAS alleles. DESIGN.— Cohort 1 consisted of 127 primary conventional PDACs with no neoadjuvant therapy, excluding colloid/mucinous, adenosquamous, undifferentiated, and intraductal papillary mucinous neoplasm-associated carcinomas, for which an in-house 42-gene mutational panel had been performed. A morphologic classification system was devised wherein each tumor was assigned as conventional, papillary/large duct (P+LD, defined as neoplastic glands with papillary structure and/or with length ≥0.5 mm), or poorly differentiated (when the aforementioned component was 60% or more of the tumor). Cohort 2 was a cohort of 88 PDACs in The Cancer Genome Atlas, which were similarly analyzed. RESULTS.— In both cohorts, there was significant enrichment of P+LD morphology in PDACs with KRAS G12V and G12R compared with G12D. In the entire combined cohort, Kaplan-Meier analyses showed longer overall survival (OS) with KRAS G12R as compared with G12D (median OS of 1255 versus 682 days, P = .03) and in patients whose PDACs displayed P+LD morphology as compared with conventional morphology (median OS of 1175 versus 684 days, P = .04). In the adjuvant-only subset, KRAS G12R had the longest OS compared with G12D, G12V, and other alleles (median OS unreached/undefined versus 1009, 1129, and 1222 days, respectively). CONCLUSIONS.— PDACs with different mutant KRAS alleles are associated with distinct morphologies and clinical outcomes, with KRAS G12R allele associated with P+LD morphology and longer OS when compared with G12D using Kaplan-Meier studies.
Collapse
Affiliation(s)
- Timothy Chao
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Zi-Xuan Wang
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Wilbur B Bowne
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
- the Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Jefferson Health, Philadelphia, Pennsylvania (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo, Jiang)
| | - Clifford J Yudkoff
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Ava Torjani
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Vishal Swaminathan
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Taylor R Kavanagh
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Austin Roadarmel
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Cyrus J Sholevar
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Shawnna Cannaday
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
- the Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Jefferson Health, Philadelphia, Pennsylvania (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo, Jiang)
| | - Geoffrey Krampitz
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
- the Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Jefferson Health, Philadelphia, Pennsylvania (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo, Jiang)
| | - Tingting Zhan
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Eliyahu Gorgov
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
- the Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Jefferson Health, Philadelphia, Pennsylvania (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo, Jiang)
| | - Avinoam Nevler
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
- the Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Jefferson Health, Philadelphia, Pennsylvania (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo, Jiang)
| | - Harish Lavu
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
- the Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Jefferson Health, Philadelphia, Pennsylvania (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo, Jiang)
| | - Charles J Yeo
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
- the Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Jefferson Health, Philadelphia, Pennsylvania (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo, Jiang)
| | - Stephen C Peiper
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Wei Jiang
- From the Department of Pathology and Genomic Medicine (Chao, Wang, Peiper, Jiang), the Department of Surgery (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo), Sidney Kimmel Medical College (Yudkoff, Torjani, Swaminathan, Kavanagh, Roadarmel, Sholevar), and the Division of Biostatistics in the Department of Pharmacology & Experimental Therapeutics (Zhan), Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
- the Pancreatic, Biliary and Related Cancer Center, Sidney Kimmel Cancer Center, Jefferson Health, Philadelphia, Pennsylvania (Bowne, Cannaday, Krampitz, Gorgov, Nevler, Lavu, Yeo, Jiang)
| |
Collapse
|
196
|
Grahovac J, Đurić A, Tanić M, Krivokuća A. Sex-Related Differences in Pancreatic Ductal Adenocarcinoma Progression and Response to Therapy. Int J Mol Sci 2024; 25:12669. [PMID: 39684385 DOI: 10.3390/ijms252312669] [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: 10/15/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly malignancies with an increasing incidence rate and limited therapeutic options. Biological sex has an impact on many aspects of PDAC development and response to therapy, yet it is highly unappreciated in both basic and translational research, and worryingly in PDAC clinical trials. In this review, we summarize how biological sex influences PDAC incidence and mortality, genetic and epigenetic landscapes, anti-tumor immunity, responses to hormones, cachexia, and the efficacy of therapy. We highlight the importance of sex as a variable and discuss how to implement it into preclinical and clinical research. These considerations should be of use to researchers aiming at improving understanding of PDAC biology and developing precision medicine therapeutic strategies.
Collapse
Affiliation(s)
- Jelena Grahovac
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Ana Đurić
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Miljana Tanić
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Ana Krivokuća
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| |
Collapse
|
197
|
Chung V, Mizrahi JD, Pant S. Novel Therapies for Pancreatic Cancer. JCO Oncol Pract 2024:OP2400279. [PMID: 39591547 DOI: 10.1200/op.24.00279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 10/15/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic adenocarcinoma (PDAC) unfortunately remains a highly fatal disease with a 5-year survival rate of only 11%. If surgical resection is not possible, systemic chemotherapy represents the standard-of-care approach to management. Combination chemotherapy regimens using fluorouracil (fluorouracil, oxaliplatin, leucovorin, and irinotecan; and fluorouracil, leucovorin, and oxaliplatin) or gemcitabine with albumin-bound paclitaxel have the potential to improve overall survival for patients with advanced disease. With the increasing understanding of the molecular drivers of pancreatic cancer, novel therapeutic approaches have made incremental progress for these patients. The molecular landscape of PDAC has been studied extensively. Approximately 90% of PDACs harbor mutations in the KRAS gene, a driver mutation that has long been considered undruggable. However, novel KRAS inhibitors have shown therapeutic promise in early-phase clinical trials, with larger studies ongoing. Less frequently encountered genomic aberrations that have therapeutic potential include NRG, BRAF, NTRK, HER2, BRCA, PALB2, and claudin. Immune checkpoint inhibitors have unfortunately yielded disappointing efficacy for the majority of patients with pancreatic cancer, except for those with tumors exhibiting deficiency in mismatch repair proteins. Alternative approaches to incorporate immunotherapy have shown more promise such as use of immune checkpoint inhibitors for selected patients in the maintenance setting and potential vaccine therapies in the postsurgery adjuvant setting. It is vital to perform molecular profiling in all patients with PDAC to identify potential treatment targets, and to enroll patients in clinical trials whenever possible.
Collapse
Affiliation(s)
- Victoria Chung
- Ochsner Medical Center, New Orleans, LA
- MD Anderson Cancer Center, Houston, TX
| | | | | |
Collapse
|
198
|
Cheng K, Li X, Lv W, Zhao G, Zhou R, Chang C, Yang H, Li R, Li Z, Chen Y, Yi C, Yan O, Xiao C, Zhang Y, Xiong J, Huang Z, Shao W, You X, Guo W, He D, Ling W, Wang R, Tian B, Zhao C, Cao D. Spatial interactions of immune cells as potential predictors to efficacy of toripalimab plus chemotherapy in locally advanced or metastatic pancreatic ductal adenocarcinoma: a phase Ib/II trial. Signal Transduct Target Ther 2024; 9:321. [PMID: 39582060 PMCID: PMC11586424 DOI: 10.1038/s41392-024-02031-8] [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: 08/12/2024] [Revised: 10/10/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
Advanced pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis. Immunotherapy alone offers limited efficacy, but it is still unknown whether its combination with chemotherapy could offer synergistic anti-tumor effects. This phase Ib/II study evaluated the safety and efficacy of combining toripalimab with the gemcitabine plus nab-paclitaxel (GnP) regimen as first-line treatment for locally advanced or metastatic PDAC and explored predictive biomarkers (ChiCTR2000032293). The primary endpoints were safety and overall survival (OS). The secondary outcomes were objective response rate (ORR), disease control rate (DCR), and progression-free survival (PFS). Immune-related biomarkers including programmed death-ligand 1 (PD-L1) expression, genetic status, cytokine levels, and spatial features of the tumor immune microenviroment (TIME) were investigated. Neither serious treatment-related adverse events nor grade 4 immune-related adverse events were reported. Among the 72 patients, the median OS was 8.9 months, 12-month OS rate was 31.9%, with median PFS of 5.6 months, ORR of 33.3%, and DCR of 90.3%. Higher PD-L1 expression, without liver metastases were associated with higher ORR, however these factors could not effectively distinguish responders and non-responders. Importantly, dendritic cells - T helper cells - cytotoxic T lymphocytes (DC-Th-CTL) enriched immune niche and their spatial interactions were dominant predictors of response based on TIME analysis using a cyclic multiplex tissue staining assay, with an area under the curve value of 0.8. Overall, GnP plus toripalimab exhibited good safety and differentiated efficacy in selected population, and the spatial interactions of DC-Th-CTL represent promising predictors to efficacy of immunochemotherapy in locally advanced or metastatic PDAC.
Collapse
Affiliation(s)
- Ke Cheng
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoying Li
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wanrui Lv
- Department of Oncology, Meishan City People's Hospital, Meishan, Sichuan, China
| | - Gang Zhao
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruihan Zhou
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University and Collaborative Innovation Center, West China Hospital, Chengdu, Sichuan, China
| | - Chen Chang
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Heqi Yang
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruizhen Li
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiping Li
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ye Chen
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Yi
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ouying Yan
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chaoxin Xiao
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University and Collaborative Innovation Center, West China Hospital, Chengdu, Sichuan, China
| | - Yi Zhang
- Pancreatic Division, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of General Surgery, ChengDu ShangJing NanFu Hospital, Chengdu, Sichuan, China
| | - Junjie Xiong
- Pancreatic Division, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zixin Huang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiology, West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | - Xin You
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenhao Guo
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Du He
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenwu Ling
- Department of Medical Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Rui Wang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bole Tian
- Pancreatic Division, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University and Collaborative Innovation Center, West China Hospital, Chengdu, Sichuan, China.
| | - Dan Cao
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
199
|
Martin RCG, White RR, Bilimoria MM, Kluger MD, Iannitti DA, Polanco PM, Hammil CW, Cleary SP, Heithaus RE, Welling T, Chan CHF. Effectiveness and Safety of Irreversible Electroporation When Used for the Ablation of Stage 3 Pancreatic Adenocarcinoma: Initial Results from the DIRECT Registry Study. Cancers (Basel) 2024; 16:3894. [PMID: 39682087 DOI: 10.3390/cancers16233894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Overall survival for patients with Stage 3 pancreatic ductal adenocarcinoma (PDAC) remains limited, with a median survival of 12 to 15 months. Irreversible electroporation (IRE) is a local tumor ablation method that induces cancerous cell death by disrupting cell membrane homeostasis. The DIRECT Registry study was designed to assess the effectiveness and safety of IRE when combined with standard of care (SOC) treatment for Stage 3 PDAC versus SOC alone in a real-world setting after at least 3 months of induction chemotherapy; Methods: Patients with Stage 3 PDAC treated with IRE plus SOC or SOC alone were prospectively enrolled in a multicenter registry study. Enrollment required 3 months of active multi-agent chemotherapy with no progression before enrollment. Endpoints were 30- and 90-day mortality and adverse events (AEs). RESULTS Eighty-seven IRE and 27 SOC subjects were enrolled in the registry. Mean ages were 64.0 ± 8.4 and 66.4 ± 9.9 years, and mean anterior/posterior tumor diameters were 2.2 ± 0.7 cm and 3.2 ± 1.3 for the IRE and SOC groups respectively (p = 0.0066). All IRE procedures were performed using an open approach. The 90-day all-cause mortality was 5/83 (6.0%) and 2/27 (7.4%) for the IRE and SOC groups, respectively. Two subjects in the IRE group died from treatment-related complications, and one patient in the SOC group died due to chemotherapy-related complications. CONCLUSIONS Initial results from the DIRECT registry study indicate the use of IRE for curative intent tumor ablation in combination with induction chemotherapy has equivalent morbidity and mortality rates when compared to standard-of-care chemotherapy alone.
Collapse
Affiliation(s)
- Robert C G Martin
- Division of Surgical Oncology, Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| | - Rebekah Ruth White
- San Diego Moores Cancer Center, University of California, La Jolla, CA 92037, USA
| | | | - Michael D Kluger
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
| | - David A Iannitti
- Atrium Health Carolinas Medical Center, Charlotte, NC 28203, USA
| | | | - Chet W Hammil
- Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Sean P Cleary
- Division of Surgical Oncology, Department of Surgery, University of Louisville, Louisville, KY 40202, USA
- Mayo Clinic, Rochester, MN 55905, USA
- University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Robert Evans Heithaus
- Department of Radiology, College of Medicine, University of South Florida, Tampa, FL 33606, USA
| | - Theodore Welling
- Division of Surgical Oncology, Department of Surgery, University of Louisville, Louisville, KY 40202, USA
- NYU Langone Health, New York, NY 10016, USA
- UC San Diego Health, San Diego, CA 92037, USA
| | - Carlos H F Chan
- University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
200
|
Tushoski-Alemán GW, Crespin AJ, Oguejiofor CJ, Szymkiewicz DD, Herremans KM, Han S, Hughes SJ. Variability of quality-of-life measurements and reporting in randomised controlled trials of pancreatic cancer: a systematic review. BMJ Open 2024; 14:e083696. [PMID: 39551595 PMCID: PMC11574412 DOI: 10.1136/bmjopen-2023-083696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 10/11/2024] [Indexed: 11/19/2024] Open
Abstract
OBJECTIVES This systematic review aims to evaluate the methodology used in pancreatic cancer (PC) randomised controlled trials (RCTs) measuring quality of life (QOL) and focuses on the type, frequency, survey compliance and duration of these assessments. DESIGN Systematic review of PC RCTs measuring QOL. DATA SOURCES A search of PubMed.gov and ClinicalTrials.gov was conducted for PC RCTs measuring QOL from inception to 21 March 2023. Only phase III RCTs were included. Studies were excluded if QOL was not measured, the study was phase I/II, in the second-line setting or unavailable in English. Data were independently extracted by two reviewers in a standardised fashion. PRIMARY AND SECONDARY OUTCOME MEASURES Primary outcomes included the type of QOL instrument used, the timing and frequency of assessments, methods of analysis and survey completion rates (SCRs) over time. Secondary outcomes included patient demographics, significant QOL improvements and the frequency of trials measuring QOL. RESULTS Out of 269 studies screened, 54 RCTs were identified, and 24 measured QOL (involving 11 229 patients). Instruments used included the EORTC QLQ-C30 (n=15), FACT-HEP (n=3), Spitzer-QOL-Index (n=2), EQ-5D (n=2), LASA (n=1) and FACT-PA (n=1). Most trials assessed QOL until disease progression or death (10/24), with 4-week intervals being the most common (7/24). SCRs were reported in 15/24 trials, with disease stage influencing SCRs over time. In trials with metastatic, locally advanced/metastatic, and resectable disease, the median times to reach a 50% response rate-defined as the point where the number of surveys completed was half of the enrolled participants-were 12.41 weeks (n=2), 14.14 weeks (n=10), and 54.2 weeks (n=3), respectively." Only 2/24 trials reported significant QOL improvements between treatment arms. Patient age was reported in all trials, while race/ethnicity was only reported in 4/24 trials. CONCLUSIONS Significant variability exists in the timing, methods and reporting of QOL assessments in PC trials. There is a need for further research to assess the implications of missing data and consider the temporality of QOL assessment in patients with advanced cancers and poor prognosis.
Collapse
Affiliation(s)
| | | | | | | | - Kelly M Herremans
- Department of Surgery, University of Florida, Gainesville, Florida, USA
| | - Song Han
- Department of Surgery, University of Florida, Gainesville, Florida, USA
| | - Steven J Hughes
- Department of Surgery, University of Florida, Gainesville, Florida, USA
| |
Collapse
|