301
|
Drougkas K, Karampinos K, Karavolias I, Gomatou G, Koumprentziotis IA, Ploumaki I, Triantafyllou E, Kotteas E. CAR-T Cell Therapy in Pancreatic and Biliary Tract Cancers: An Updated Review of Clinical Trials. J Gastrointest Cancer 2024; 55:990-1003. [PMID: 38695995 DOI: 10.1007/s12029-024-01054-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Pancreatic and biliary tract cancers are digestive system tumors with dismal prognosis and limited treatment options. The effectiveness of conventional surgical interventions, radiation therapy, and systemic therapy is restricted in these cases. Furthermore, clinical trials have shown that immunotherapy using immune checkpoint inhibitors has only demonstrated modest clinical results when applied to patients with pancreatobiliary tumors. This highlights the importance of implementing combination immunotherapy approaches or exploring alternative therapeutic strategies to improve treatment outcomes. MATERIALS AND METHODS We reviewed the relevant literature on chimeric antigen receptor (CAR)-T cell therapy for pancreatobiliary cancers from PubMed/Medline and ClinicalTrials.gov and retrieved the relevant data accordingly. Attention was additionally given to the examination of grey literature with the aim of obtaining additional details regarding ongoing clinical trials. We mainly focused on abstracts and presentations and e-posters and slides of recent important annual meetings (namely ESMO Immuno-Oncology Congress, ESMO Congress, ASCO Virtual Scientific Program, ASCO Gastrointestinal Cancers Symposium). RESULTS CAR-T cell therapy has emerged as a promising and evolving treatment approach for pancreatic and biliary tract cancer. This form of adoptive cell therapy utilizes genetic engineering to modify the expression of specific antibodies on the surface of T cells enabling them to target specific cancer-associated antigens and to induce potent anti-tumor activity. The aim of this review is to provide an updated summary of the available evidence from clinical trials that have explored the application of CAR-T cell therapy in treating pancreatobiliary cancers. CONCLUSIONS While the utilization of CAR-T cell therapy in pancreatobiliary cancers is still in its initial phases with only a limited amount of clinical data available, the field is advancing rapidly, incorporating novel technologies to mitigate potential toxicities and enhance antigen-directed tumor eradication.
Collapse
Affiliation(s)
- Konstantinos Drougkas
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Karampinos
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Karavolias
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Gomatou
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis-Alexios Koumprentziotis
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | - Ioanna Ploumaki
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthymios Triantafyllou
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Elias Kotteas
- Oncology Unit, 3rd Department of Medicine, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
302
|
Huerta M, Martín-Arana J, Gimeno-Valiente F, Carbonell-Asins JA, García-Micó B, Martínez-Castedo B, Robledo-Yagüe F, Camblor DG, Fleitas T, García Bartolomé M, Alfaro-Cervelló C, Garcés-Albir M, Dorcaratto D, Muñoz-Forner E, Seguí V, Mora-Oliver I, Gambardella V, Roselló S, Sabater L, Roda D, Cervantes A, Tarazona N. ctDNA whole exome sequencing in pancreatic ductal adenocarcinoma unveils organ-dependent metastatic mechanisms and identifies actionable alterations in fast progressing patients. Transl Res 2024; 271:105-115. [PMID: 38782356 DOI: 10.1016/j.trsl.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
Understanding progression mechanisms and developing new targeted therapies is imperative in pancreatic ductal adenocarcinoma (PDAC). In this study, 80 metastatic PDAC patients were prospectively recruited and divided into discovery (n=37) and validation (n=43) cohorts. Tumor and plasma samples taken at diagnosis were pair analyzed using whole exome sequencing (WES) in patients belonging to the discovery cohort alone. The variant allele frequency (VAF) of KRAS mutations was measured by ddPCR in plasma at baseline and response assessment in all patients. Plasma WES identified at least one pathogenic variant across the cohort, uncovering oncogenic mechanisms, DNA repair, microsatellite instability, and alterations in the TGFb pathway. Interestingly, actionable mutations were mostly found in plasma rather than tissue. Patients with shorter survival showed enrichment in cellular organization regulatory pathways. Through WES we could identify a specific molecular profile of patients with liver metastasis, which exhibited exclusive mutations in genes related to the adaptive immune response pathway, highlighting the importance of the immune system in liver metastasis development. Moreover, KRAS mutations in plasma (both at diagnosis and persistent at follow-up) correlated with shorter progression free survival (PFS). Patients presenting a reduction of over 84.75 % in KRAS VAF at response assessment had similar PFS to KRAS-negative patients. Overall, plasma WES reveals molecular profiles indicative of rapid progression, potentially actionable targets, and associations between adaptive immune response pathway alterations and liver tropism.
Collapse
Affiliation(s)
- Marisol Huerta
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Jorge Martín-Arana
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Gimeno-Valiente
- Cancer Evolution and Genome Instability Laboratory, University College London Cancer Institute, London, UK
| | | | - Blanca García-Micó
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Belén Martínez-Castedo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Fabián Robledo-Yagüe
- Bioinformatics Unit, INCLIVA Biomedical Research Institute, University of Valencia, Spain
| | - Daniel G Camblor
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Tania Fleitas
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel García Bartolomé
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Clara Alfaro-Cervelló
- Department of Pathology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Marina Garcés-Albir
- Liver, Biliary and Pancreatic Unit, Department of General Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario de Valencia, University of Valencia, Valencia, Spain
| | - Dimitri Dorcaratto
- Liver, Biliary and Pancreatic Unit, Department of General Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario de Valencia, University of Valencia, Valencia, Spain
| | - Elena Muñoz-Forner
- Liver, Biliary and Pancreatic Unit, Department of General Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario de Valencia, University of Valencia, Valencia, Spain
| | - Víctor Seguí
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Isabel Mora-Oliver
- Liver, Biliary and Pancreatic Unit, Department of General Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario de Valencia, University of Valencia, Valencia, Spain
| | - Valentina Gambardella
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Susana Roselló
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Luis Sabater
- Liver, Biliary and Pancreatic Unit, Department of General Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario de Valencia, University of Valencia, Valencia, Spain
| | - Desamparados Roda
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Andrés Cervantes
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| | - Noelia Tarazona
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
303
|
Marcinak CT, Ahmed KS, LoConte NK, Praska CE, Varley PR, Weber SM, Abbott DE, Ronnekleiv-Kelly SM, Kratz JD, Lubner SJ, Minter RM, Zafar SN. Understanding futility in pancreaticoduodenectomy: Insights from a national cohort. J Surg Oncol 2024; 130:462-475. [PMID: 39082628 DOI: 10.1002/jso.27773] [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: 06/14/2024] [Accepted: 06/23/2024] [Indexed: 10/16/2024]
Abstract
BACKGROUND AND OBJECTIVES Pancreaticoduodenectomy (PD), the only surgical option for right-sided pancreatic ductal adenocarcinoma (PDAC), carries significant morbidity. Not all patients may be deriving a survival benefit from this operation. We sought to identify the rate of futile PD and its associated factors in a large national cohort. METHODS We performed a retrospective analysis using the National Cancer Database (2004-2020), including all patients who underwent PD for non-metastatic PDAC. The primary outcome was operative futility, which was defined as death within 12 months of diagnosis despite PD. Multivariable regression was used to identify factors associated with futility. We performed a subgroup analysis on patients who received neoadjuvant systemic therapy. RESULTS Data from 66 326 patients were analyzed, and 16 772 (25.3%) underwent PD that met criteria for futility. Macroscopically positive margins (odds ratio [OR]: 2.87; 95% confidence interval [CI]: 2.36-3.48), poor tumor differentiation (OR: 2.44; 95% CI: 2.25-2.65), and N2 nodal stage (OR: 2.09; 95% CI: 1.98-2.20) were associated with the greatest odds of futility. Meanwhile, receipt of any systemic therapy (OR: 0.33; 95% CI: 0.31-0.34), receipt of any radiation (OR: 0.60; 95% CI: 0.57-0.63), and receipt of neoadjuvant systemic therapy (OR: 0.62; 95% CI: 0.57-0.66) were associated with the lowest odds of futility. In the neoadjuvant subgroup, a longer diagnosis-to-surgery interval was associated with lower odds of futility. CONCLUSION PD was futile in about one quarter of patients. Futility was associated with higher age and worse tumor biology. Receipt of neoadjuvant therapy resulted in fewer futile operations.
Collapse
Affiliation(s)
- Clayton T Marcinak
- Division of Surgical Oncology, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kaleem S Ahmed
- Division of Surgical Oncology, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Noelle K LoConte
- Division of Hematology, Oncology, and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Corinne E Praska
- Division of Surgical Oncology, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Patrick R Varley
- Division of Surgical Oncology, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Daniel E Abbott
- Division of Surgical Oncology, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sean M Ronnekleiv-Kelly
- Division of Surgical Oncology, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jeremy D Kratz
- Division of Hematology, Oncology, and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sam J Lubner
- Division of Hematology, Oncology, and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Rebecca M Minter
- Division of Surgical Oncology, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Syed Nabeel Zafar
- Division of Surgical Oncology, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
304
|
Yim K, Seo KJ, Abdul-Ghafar J, Alam MR, Paik KY, Chong Y, Shin OR. Poly (Adp-Ribose) Polymerase-1 (PARP-1) Is a Good Prognostic Marker for Pancreatic/Periampullary Cancers. Pancreas 2024; 53:e681-e688. [PMID: 38530967 DOI: 10.1097/mpa.0000000000002356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
BACKGROUND Periampullary cancer (PAC) is highly aggressive with no effective adjuvant therapy or prognostic markers. Recently, poly (ADP-ribose) polymerase-1 (PARP-1) has emerged as a target in solid cancers, and its relationship with epithelial-mesenchymal transition (EMT) has been observed. However, the relationship between PARP-1 and EMT in PAC has not explored well. MATERIALS AND METHODS We assessed the prognostic significance of PARP-1 in 190 PACs patients and correlated it with EMT markers, including FGF8, FGFR4, MMP2, MMP3, Snail, and ZEB1. Immunohistochemistry for PARP-1 and EMT markers was performed using a tissue microarray. RESULTS PARP-1 and FGF8 expression were associated with better survival unlike other solid cancers ( P = 0.006 and P = 0.003), and MMP3 and ZEB1 expression were associated with poor prognosis in multivariate and survival analyses ( P = 0.009 and P < 0.001). In addition, PARP-1 is related negatively to Snail but not related with other EMT markers, implying an independent mechanism between PARP-1 and EMT in PACs. PARP-1 and FGF8 are independent good survival markers in PACs unlike other solid cancers. CONCLUSIONS PARP-1 and FGF8 in PACs could not be related to the EMT pathway but must be rather understood in light of similar cancer-protective roles. Further studies are required on EMT-associated immune markers in PACs.
Collapse
Affiliation(s)
| | | | | | | | - Kwang Yeol Paik
- Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | | |
Collapse
|
305
|
Adenis A, Ghiringhelli F, Gauthier L, Mazard T, Evesque L, Evrard A, Chalbos P, Moussion A, Gourgou S, Ychou M. Regorafenib plus FOLFIRINOX as first-line treatment for patients with RAS-mutant metastatic colorectal cancer (FOLFIRINOX-R trial): a dose-escalation study. Cancer Chemother Pharmacol 2024; 94:443-452. [PMID: 38987363 PMCID: PMC11420384 DOI: 10.1007/s00280-024-04682-4] [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: 02/08/2024] [Accepted: 05/29/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE The combination of bevacizumab and FOLFIRINOX is used in patients with RAS-mutant metastatic colorectal cancer (RASm-mCRC). Regorafenib, an oral multi-tyrosine kinase inhibitor, has antiangiogenic properties, cytostatic effects and also true cytotoxic effects, unlike bevacizumab. The aim of this study was to determine the maximum tolerated dose (MTD) and the recommended phase 2 dose (RP2D) of the regorafenib-FOLFIRINOX combination in patients with RASm-mCRC. METHODS The FOLFIRINOX-R trial was a phase 1/2 study where the dose-escalation part (3 + 3 design with three dose levels, DLs) was completed before its early termination. FOLFIRINOX (14-day cycle) included oxaliplatin (standard dose), folinic acid, fluorouracil and irinotecan (150 or 180 mg/m²). Regorafenib (120 or 160 mg daily) was given from day 4 to day 10 of each cycle. Dose-limiting toxicity (DLT) was studied in the first three cycles. Eligibility criteria included ECOG performance status ≤ 1 and not previously treated RASm-mCRC. RESULTS Thirteen patients (median age: 65 years; min-max: 40-76) were enrolled. DLT could not be evaluated in one patient (DL3) due to poor observance. The median treatment duration and median follow-up were 6.2 (min-max: 2.3-10) and 13.4 (min-max: 3.8-18.0) months, respectively. Dose was modified in 12/13 (92%) patients. One grade 3 hypokalemia occurred at DL2. MTD was not reached at DL3. Grade 3 diarrhea was recorded in 7/13 patients (13 events) equally distributed in all DLs. CONCLUSION The RP2D for this regorafenib-FFX combination could not be determined due to a high prevalence of grade 3 diarrhea related to treatment as advised by our Independent Data Monitoring Committee. TRIAL REGISTRATION NUMBERS ClinicalTrials.gov : NCT03828799.
Collapse
Affiliation(s)
- Antoine Adenis
- Medical Oncology Department, Montpellier Cancer Institute (ICM), INSERM U1194, Montpellier University, Montpellier, 34298, France.
| | | | - Ludovic Gauthier
- Biometrics Unit, Montpellier Cancer Institute (ICM), Montpellier University, Montpellier, France
| | - Thibault Mazard
- Medical Oncology Department, Montpellier Cancer Institute (ICM), INSERM U1194, Montpellier University, Montpellier, 34298, France
| | - Ludovic Evesque
- Medical Oncology Department, Centre Antoine Lacassagne, Nice, France
| | - Alexandre Evrard
- Montpellier Cancer Research Institute (IRCM), INSERM U1194, Montpellier University, Montpellier, France
- Laboratory of Biochemistry and Molecular Biology, Nimes University Hospital, IRCM, University of Montpellier, INSERM, Montpellier, France
| | - Patrick Chalbos
- Clinical Research and Innovation Department, Montpellier Cancer Institute (ICM), Montpellier University, Montpellier, France
| | - Aurore Moussion
- Clinical Research and Innovation Department, Montpellier Cancer Institute (ICM), Montpellier University, Montpellier, France
| | - Sophie Gourgou
- Biometrics Unit, Montpellier Cancer Institute (ICM), Montpellier University, Montpellier, France
| | - Marc Ychou
- Medical Oncology Department, Montpellier Cancer Institute (ICM), INSERM U1194, Montpellier University, Montpellier, 34298, France
| |
Collapse
|
306
|
Niwa H, Nakamura T, Kushiya H, Kuraya T, Inoko K, Inagaki A, Suzuki T, Sasaki K, Tsuchikawa T, Hiraoka K, Shichinohe T, Hatanaka Y, Jolly DJ, Kasahara N, Hirano S. Therapeutic activity of retroviral replicating vector-mediated gene therapy in combination with anti-PD-1 antibody in a murine pancreatic cancer model. Cancer Gene Ther 2024; 31:1390-1401. [PMID: 39039195 DOI: 10.1038/s41417-024-00810-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 05/24/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024]
Abstract
Toca 511, a tumor-selective retroviral replicating vector encoding the yeast cytosine deaminase (yCD) gene, exerts direct antitumor effects through intratumoral prodrug 5-fluorocytosine (5-FC) conversion to active drug 5-fluorouracil by yCD, and has demonstrated therapeutic efficacy in preclinical and clinical trials of various cancers. Toca 511/5-FC treatment may also induce antitumor immunity. Here, we first examined antitumor immune responses activated by Toca 511/5-FC treatment in an immunocompetent murine pancreatic cancer model. We then evaluated the therapeutic effects achieved in combination with anti-programmed cell death protein 1 antibody. In the bilateral subcutaneous tumor model, as compared with the control group, enhanced CD8+ T-cell-mediated cytotoxicity and increased T-cell infiltration in Toca 511-untransduced contralateral tumors were observed. Furthermore, the expression levels of T-cell co-inhibitory receptors on CD8+ T-cells increased during treatment. In the bilateral subcutaneous tumor model, combination therapy showed significantly stronger tumor growth inhibition than that achieved with either monotherapy. In an orthotopic tumor and peritoneal dissemination model, the combination therapy resulted in complete regression in both transduced orthotopic tumors and untransduced peritoneal dissemination. Thus, Toca 511/5-FC treatment induced a systemic antitumor immune response, and the combination therapy could be a promising clinical strategy for treating metastatic pancreatic cancer.
Collapse
Affiliation(s)
- Hiroki Niwa
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
| | - Toru Nakamura
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan.
| | - Hiroki Kushiya
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
| | - Tomotaka Kuraya
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
| | - Kazuho Inoko
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
| | - Akihito Inagaki
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Tomohiro Suzuki
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
| | - Katsunori Sasaki
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
| | - Takahiro Tsuchikawa
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
| | - Kei Hiraoka
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
- Department of Clinical Research, NHO Hakodate National Hospital, Hakodate, Hokkaido, Japan
| | - Toshiaki Shichinohe
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
| | - Yutaka Hatanaka
- Center for Development of Advanced Diagnostics (C-DAD), Hokkaido University Hospital, Sapporo, Japan
| | - Douglas J Jolly
- Tocagen Inc., San Diego, CA, USA
- Abintus Bio Inc., San Diego, CA, USA
| | - Noriyuki Kasahara
- Department of Neurological Surgery, University of California, San Francisco, CA, USA.
- Department of Radiation Oncology, University of California, San Francisco, CA, USA.
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
| |
Collapse
|
307
|
Yun WG, Han Y, Jung HS, Kwon W, Park JS, Jang JY. Emerging role of local treatment in the era of advanced systemic treatment in pancreatic cancer with liver metastasis: A systematic review and meta-analysis. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:601-610. [PMID: 39020260 DOI: 10.1002/jhbp.12051] [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: 07/19/2024]
Abstract
BACKGROUND Approximately 50% of pancreatic cancer cases are diagnosed with distant metastases, commonly in the liver, leading to poor prognosis. With modern chemotherapy regimens extending patient survival and stabilizing metastasis, there has been a rise in the use of local treatments. However, the effectiveness for local treatment remains unclear. METHODS PubMed, Embase, and Cochrane databases were searched for studies reporting the survival outcomes of pancreatic cancer cases with isolated synchronous or metachronous liver metastases who underwent curative-intent local treatment. Hazard ratios were combined using a random-effects model. RESULTS The full texts of 102 studies were screened, and 14 retrospective studies were included in the meta-analysis. Among patients with synchronous liver metastases, overall survival was significantly better in those who underwent curative-intent local treatment than in those who did not (hazard ratio [HR]: 0.35, 95% confidence interval [CI]: 0.24-0.52). Among patients with metachronous liver metastases, overall survival was also significantly better in those who underwent curative-intent local treatment than in those who did not (HR 0.37, 95% CI: 0.19-0.73). CONCLUSIONS Curative-intent local treatment may be a feasible option for highly selected pancreatic cancer cases with liver metastases. However, the optimal strategy for local treatments should be explored in future studies.
Collapse
Affiliation(s)
- Won-Gun Yun
- Department of Surgery, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Youngmin Han
- Department of Surgery, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hye-Sol Jung
- Department of Surgery, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wooil Kwon
- Department of Surgery, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joon Seong Park
- Department of Surgery, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Young Jang
- Department of Surgery, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
308
|
Chen BS, Chan SY, Bteich F, Kuang C, Meyerhardt JA, Ma KSK. Safety and efficacy of liposomal irinotecan as the second-line treatment for advanced pancreatic cancer: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2024; 201:104386. [PMID: 38735505 DOI: 10.1016/j.critrevonc.2024.104386] [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: 12/05/2023] [Revised: 04/09/2024] [Accepted: 05/05/2024] [Indexed: 05/14/2024] Open
Abstract
INTRODUCTION Nanoliposomal irinotecan (nal-IRI) is a novel regimen for pancreatic cancer, featuring a longer half-life and an increased area under the concentration-time curve. This study aims to assess the safety and efficacy of nal-IRI as a second-line treatment for advanced pancreatic cancer. METHODS A systemic literature search was conducted based on articles published before September 26th, 2023 in databases, including PubMed, Cochrane Library, EMBASE and Web of Science. The fixed effects model was used to calculate the pooled mean difference for overall survival (OS) and progression-free survival (PFS), as well as the pooled odds ratio for the overall response rate (ORR) and the risk of adverse events. RESULTS A total of 21 studies, including 3044 patients with locally advanced unresectable or metastatic pancreatic cancers, were considered eligible. The use of nal-IRI, combined with 5-fluorouracil and leucovorin, resulted in significantly improved PFS (pooled mean difference=1.01 months, 95 % confidence interval [CI]=0.97-1.05, p<0.01) and OS (pooled mean difference=0.29 months, 95 %CI=0.18-0.39, p<0.01), as well as significantly better ORR (pooled odds ratio=2.06, 95 %CI=1.30-3.27, p=0.002) compared to other second-line regimens. Nonetheless, an increased risk of grade 3 or greater neutropenia, anemia, hypokalemia, diarrhea, and vomiting was also noted. CONCLUSION Second-line treatments based on nal-IRI exhibited significantly improved PFS, OS, and ORR compared to other available treatments in advanced pancreatic cancer. Further research is necessary to corroborate these findings and define the role of nal-IRI in both first and later lines of therapy.
Collapse
Affiliation(s)
- Brian Shiian Chen
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shu-Yen Chan
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fernand Bteich
- Montefiore Einstein Comprehensive Cancer Center, Bronx, NY, USA; Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Chaoyuan Kuang
- Montefiore Einstein Comprehensive Cancer Center, Bronx, NY, USA; Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jeffery A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Kevin Sheng-Kai Ma
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
309
|
Keane F, Chou JF, Walch H, Schoenfeld J, Singhal A, Cowzer D, Harrold E, O’Connor CA, Park W, Varghese A, El Dika I, Balogun F, Yu KH, Capanu M, Schultz N, Yaeger R, O’Reilly EM. Precision medicine for pancreatic cancer: characterizing the clinicogenomic landscape and outcomes of KRAS G12C-mutated disease. J Natl Cancer Inst 2024; 116:1429-1438. [PMID: 38702822 PMCID: PMC11378314 DOI: 10.1093/jnci/djae095] [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: 01/10/2024] [Revised: 03/27/2024] [Accepted: 04/20/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Mutated Kirsten rat sarcoma viral oncogene homolog (KRAS) is the most common oncogene alteration in pancreatic ductal adenocarcinoma, and KRAS glycine to cystine substitution at codon 12 (G12C) mutations (KRAS G12Cmut) are observed in 1%-2%. Several inhibitors of KRAS G12C have recently demonstrated promise in solid tumors, including pancreatic cancer. Little is known regarding clinical, genomics, and outcome data of this population. METHODS Patients with pancreatic cancer and KRAS G12Cmut were identified at Memorial Sloan Kettering Cancer Center and via the American Association of Cancer Research Project Genomics, Evidence, Neoplasia, Information, Exchange database. Clinical, treatment, genomic, and outcomes data were analyzed. A cohort of patients at Memorial Sloan Kettering Cancer Center with non-G12C KRAS pancreatic cancer was included for comparison. RESULTS Among 3571 patients with pancreatic ductal adenocarcinoma, 39 (1.1%) with KRAS G12Cmut were identified. Median age was 67 years, and 56% were female. Median body mass index was 29.2 kg/m2, and 67% had a smoking history. Median overall survival was 13 months (95% CI: 9.4 months, not reached) for stage IV and 26 months (95% CI: 23 months, not reached) for stage I-III. Complete genomic data (via American Association of Cancer Research Project Genomics, Evidence, Neoplasia, Information, Exchange database) was available for 74 patients. Most common co-alterations included TP53 (73%), CDKN2A (33%), SMAD4 (28%), and ARID1A (21%). Compared with a large cohort (n = 2931) of non-G12C KRAS-mutated pancreatic ductal adenocarcinoma, ARID1A co-mutations were more frequent in KRAS G12Cmut (P < .05). Overall survival did not differ between KRAS G12Cmut and non-G12C KRAS pancreatic ductal adenocarcinoma. Germline pathogenic variants were identified in 17% of patients; 2 patients received KRAS G12C-directed therapy. CONCLUSION Pancreatic cancer and KRAS G12Cmut may be associated with a distinct clinical phenotype. Genomic features are similar to non-G12C KRAS-mutated pancreatic cancer, although enrichment of ARID1A co-mutations was observed. Targeting of KRAS G12C in pancreatic cancer provides a precedent for broader KRAS targeting in pancreatic cancer.
Collapse
Affiliation(s)
- Fergus Keane
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joanne F Chou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Henry Walch
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joshua Schoenfeld
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anupriya Singhal
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Darren Cowzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emily Harrold
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Catherine A O’Connor
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Anna Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Imane El Dika
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Fiyinfolu Balogun
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Kenneth H Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Eileen M O’Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
310
|
Tezuka S, Ozaka M, Furuse J, Yokoyama M, Uemura K, Sano Y, Nakachi K, Imaoka H, Unno M, Shirakawa H, Shimizu S, Kato N, Kojima Y, Sano K, Kobayashi S, Terashima T, Morizane C, Ikeda M, Ueno M. Early tumor shrinkage as a prognostic predictor in chemotherapy-naïve patients with locally advanced pancreatic cancer treated with modified FOLFIRINOX or gemcitabine plus nab-paclitaxel combination therapy: An exploratory analysis of JCOG1407. Pancreatology 2024; 24:909-916. [PMID: 39060124 DOI: 10.1016/j.pan.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/04/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Early tumor shrinkage (ETS) is a prognostic predictor for patients treated with chemotherapy in colorectal cancer, although scarce studies evaluated its potential in locally advanced pancreatic cancer (LAPC). In this exploratory analysis of JCOG1407, a randomized phase II study comparing modified 5-fluorouracil, levofolinate, irinotecan, and oxaliplatin (mFOLFIRINOX) and gemcitabine plus nab-paclitaxel (GnP), we evaluated whether ETS can predict prognosis of patients with LAPC. METHODS Of the 126 patients enrolled in JCOG1407, 112 with measurable lesions were included in this study. ETS was defined as a ≥20 % reduction in tumor diameter compared with baseline at the initial imaging assessment 6-10 weeks after initiating chemotherapy. Patients were divided into the ETS (achieved ETS) and non-ETS (failed to achieve ETS) groups based on their ETS status. The impact of ETS on overall survival (OS) was compared using multivariable Cox regression analysis. RESULTS Fourteen of 55 (25.5 %) and 24 of 57 (42.1 %) patients in the mFOLFIRINOX and GnP arms, respectively, achieved ETS. In the overall population, mFOLFIRINOX arm, and GnP arm, the median OS in the ETS and non-ETS groups was 27.1 and 20.4, 29.8 and 20.6, and 24.1 and 20.4, months, respectively. The adjusted hazard ratios of OS for the ETS group in the overall population, mFOLFIRINOX arm, and GnP arm were 0.451 (95 % confidence interval [CI]: 0.270-0.754), 0.371 (95 % CI: 0.149-0.926), and 0.508 (95 % CI: 0.255-1.004), respectively. CONCLUSIONS ETS may be a prognostic predictor in chemotherapy-naïve patients with LAPC treated with mFOLFIRINOX or GnP.
Collapse
Affiliation(s)
- Shun Tezuka
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan.
| | - Masato Ozaka
- Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Masayuki Yokoyama
- JCOG Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Kohei Uemura
- JCOG Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Yusuke Sano
- JCOG Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Kohei Nakachi
- Department of Medical Oncology, Tochigi Cancer Center, Utsunomiya, Japan
| | - Hiroshi Imaoka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hirofumi Shirakawa
- Department of Medical Oncology, Tochigi Cancer Center, Utsunomiya, Japan
| | - Satoshi Shimizu
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Naoya Kato
- Department of Gastroenterology, Chiba University, Chiba, Japan
| | - Yasushi Kojima
- Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Keiji Sano
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Satoshi Kobayashi
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Takeshi Terashima
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
311
|
Lee M, Chae YS, Park S, Yun WG, Jung HS, Han Y, Kwon W, Park JS, Jang JY. Re-evaluation of risk and oncological outcomes of resection of veins and arteries in the resection of pancreatic cancer. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:671-679. [PMID: 39004799 DOI: 10.1002/jhbp.12048] [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: 07/16/2024]
Abstract
BACKGROUND Advances in chemotherapy have led to increasing major vascular resection during pancreatectomy which has been contraindicated due to high morbidity. This study aimed to verify the safety and oncological outcomes of vascular resection during pancreatectomy in the era of neoadjuvant therapy. METHODS Data from patients who underwent surgery for pancreatic cancer at Seoul National University Hospital between 2001 and 2021 were reviewed. Clinicopathological outcomes were analyzed according vessel resection. A propensity-score-matched (PSM) analysis was performed to evaluate survival outcomes. RESULTS Of 1596 patients, the proportion of those who underwent vascular resection increased from 9.2% to 23.4% over time divided into 5-year intervals. There were no differences in major complications (15.6% vs. 13.0%; p = .266) and 30-day mortality rate (0.3% vs. 0.6%; p = .837) between the vascular and nonvascular resection groups. After PSM, the vascular resection group demonstrated comparable survival outcome with the nonvascular resection group (5 year-survival-rate 20.4 vs. 23.7%; p = .194). Arterial resection yielded comparable survival outcome with nonvascular resection (5 year-survival-rate 38.1% vs. 23.7%; p = .138). CONCLUSIONS Appropriate vascular resection-even arterial-is safe and effective in patients carefully selected for radical surgery in the era of neoadjuvant therapy. Further studies are needed to determine the optimal indication and method for vascular resection in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Mirang Lee
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, Seoul, Korea
| | - Yoon Soo Chae
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seulah Park
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Won-Gun Yun
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hye-Sol Jung
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Joon Seong Park
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
312
|
Shaikh B, Gul A, Singh A, Irfan H, Ali T, Karamat R, Akilimali A. A rare case of primary signet-ring adenocarcinoma of anorectal region in a young patient: Diagnostic challenges and therapeutic outcomes. Clin Case Rep 2024; 12:e9422. [PMID: 39253370 PMCID: PMC11381185 DOI: 10.1002/ccr3.9422] [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: 05/24/2024] [Revised: 07/24/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
Key Clinical Message Primary signet-ring cell carcinoma of the anal canal and rectum is an extremely rare and aggressive malignancy. The present case underscores the importance of considering primary signet-ring cell carcinoma in differential diagnoses for young patients with chronic anorectal symptoms. It highlights the need for a multidisciplinary treatment approach (including surgery, chemotherapy, and radiotherapy) and comprehensive follow-up for managing this challenging condition and improving long-term patient outcomes. Abstract Primary signet-ring cell carcinoma of the anal canal and rectum is an exceedingly rare subtype of colorectal adenocarcinoma, often originating as an extension of rectal adenocarcinoma. This malignancy constitutes a small fraction of colorectal cancers and is scarcely reported in medical literature. We present the case of an 18-year-old male with a three-year history of progressively worsening hematochezia, anorectal pain, and defecation-associated prolapse. Initial conservative treatments failed, leading to further investigations that revealed a palpable, nodular anorectal mass. Imaging studies (including CT and MRI), and biopsy confirmed poorly differentiated adenocarcinoma with signet-ring cell morphology. The tumor exhibited extensive lymphovascular invasion and involved perirectal lymph nodes, and was staged as pT3, N2a. Immunohistochemical staining was positive for CK 7, CK 20, and SATB2, supporting the primary anorectal origin. The treatment regimen included initial diversion colostomies for symptom relief, followed by neoadjuvant chemotherapy with a modified 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) regimen and concurrent chemoradiation with Xeloda. The patient subsequently underwent an abdominoperineal resection (APR), which confirmed the diagnosis and achieved curative resection. Postoperative complications included transient ileus and wound infection, which were managed with supportive care. This case underscores the diagnostic and therapeutic challenges posed by primary signet-ring cell carcinoma of the anorectal region, highlighting the need for a high index of suspicion and comprehensive diagnostic workup in atypical presentations. The multimodal treatment approach, incorporating surgery, chemotherapy, and radiotherapy, was crucial in managing this locally advanced tumor. The rarity and aggressiveness of this carcinoma necessitate a tailored treatment strategy to improve patient outcomes. Long-term follow-up, including regular imaging and surveillance, is vital for monitoring disease recurrence and evaluating treatment effectiveness.
Collapse
Affiliation(s)
- Bisma Shaikh
- Department of Internal Medicine Jinnah Sindh Medical University Karachi Pakistan
| | - Areeba Gul
- Department of Internal Medicine Jinnah Sindh Medical University Karachi Pakistan
| | - Ajeet Singh
- Department of Internal Medicine Dow University of Health Sciences Karachi Pakistan
| | - Hamza Irfan
- Department of Medicine Shaikh Khalifa Bin Zayed Al Nahyan Medical and Dental College Lahore Pakistan
| | - Tooba Ali
- Department of Internal Medicine Dow University of Health Sciences Karachi Pakistan
| | - Riyan Karamat
- Department of Internal Medicine Rahbar Medical and Dental College Lahore Pakistan
| | - Aymar Akilimali
- Faculty of Medicine Official University of Bukavu Bukavu Democratic Republic of Congo
| |
Collapse
|
313
|
Theocharopoulos C, Ziogas IA, Douligeris CC, Efstathiou A, Kolorizos E, Ziogas DC, Kontis E. Antibody-drug conjugates for hepato-pancreato-biliary malignancies: "Magic bullets" to the rescue? Cancer Treat Rev 2024; 129:102806. [PMID: 39094332 DOI: 10.1016/j.ctrv.2024.102806] [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: 05/19/2024] [Revised: 07/17/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Hepato-Pancreato-Biliary (HPB) malignancies constitute a highly aggressive group of cancers that have a dismal prognosis. Patients not amenable to curative intent surgical resection are managed with systemic chemotherapy which, however, confers little survival benefit. Antibody-Drug Conjugates (ADCs) are tripartite compounds that merge the intricate selectivity and specificity of monoclonal antibodies with the cytodestructive potency of attached supertoxic payloads. In view of the unmet need for drugs that will enhance the survival rates of HPB cancer patients, the assessment of ADCs for treating HPB malignancies has become the focus of extensive clinical and preclinical investigation, showing encouraging preliminary results. In the current review, we offer a comprehensive overview of the growing body of evidence on ADC approaches tested for HPB malignancies. Starting from a concise discussion of the functional principles of ADCs, we summarize here all available data from preclinical and clinical studies evaluating ADCs in HPB cancers.
Collapse
Affiliation(s)
| | - Ioannis A Ziogas
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | | | | | | | - Dimitrios C Ziogas
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens 11527, Greece
| | - Elissaios Kontis
- Department of Surgery, Metaxa Cancer Hospital, Piraeus 18537, Greece
| |
Collapse
|
314
|
Gutierrez-Sainz L, Heredia-Soto V, Rodríguez-García AM, Crespo Sánchez MG, Serrano-Olmedo MG, Molero-Luis M, Losantos-García I, Ghanem I, Pérez-Wert P, Custodio A, Mendiola M, Feliu J. Cytokines and Pancreatic Ductal Adenocarcinoma: Exploring Their Relationship with Molecular Subtypes and Prognosis. Int J Mol Sci 2024; 25:9368. [PMID: 39273323 PMCID: PMC11395259 DOI: 10.3390/ijms25179368] [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: 07/20/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by its poor prognosis. The current challenge remains the absence of predictive biomarkers. Cytokines are crucial factors in the pathogenesis and prognosis of PDAC. Furthermore, there is growing interest in differentiating between molecular subtypes of PDAC. The aim of our study is to evaluate the association between the analyzed cytokines and the molecular subtypes of PDAC and to determine their prognostic value. Cytokine levels were measured in 73 patients, and molecular subtypes were analyzed in 34 of these patients. Transforming Growth Factor Beta 2 (TGF-β2) levels were independently associated with the basal-like and null subtypes. In patients with locally advanced and metastatic PDAC, elevated levels of interleukin (IL)-1α, IL-1β, IL-6, IL-8, IL-9, and IL-15 were associated with a higher risk of progression during first-line treatment, and increased levels of IL-1β, IL-6, IL-8, IL-9, and IL-15 were related to increased mortality. Furthermore, a significant association was observed between higher percentiles of IL-6 and IL-8 and shorter progression-free survival (PFS) during first-line treatment, and between higher percentiles of IL-8 and shorter overall survival (OS). In the multivariate analysis, only elevated levels of IL-8 were independently associated with a higher risk of progression during first-line treatment and mortality. In conclusion, the results of our study suggest that cytokine expression varies according to the molecular subtype of PDAC and that cytokines also play a relevant role in patient prognosis.
Collapse
Affiliation(s)
- Laura Gutierrez-Sainz
- Medical Oncology Department, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Victoria Heredia-Soto
- Translational Oncology Research Laboratory, Biomedical Research Institute, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029 Madrid, Spain
| | | | - María Gema Crespo Sánchez
- Clinical Analysis Department, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - María Gemma Serrano-Olmedo
- Clinical Analysis Department, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Marta Molero-Luis
- Clinical Analysis Department, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Itsaso Losantos-García
- Biostatistics Department, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Ismael Ghanem
- Medical Oncology Department, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Pablo Pérez-Wert
- Medical Oncology Department, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Ana Custodio
- Medical Oncology Department, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Marta Mendiola
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029 Madrid, Spain
- Molecular Pathology and Therapeutic Targets Lab, Pathology Department, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Jaime Feliu
- Medical Oncology Department, La Paz University Hospital, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
- Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), 28029 Madrid, Spain
- Cátedra UAM-AMGEN, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
315
|
Chen K, Li T, Diao H, Wang Q, Zhou X, Huang Z, Wang M, Mao Z, Yang Y, Yu W. SIRT7 knockdown promotes gemcitabine sensitivity of pancreatic cancer cell via upregulation of GLUT3 expression. Cancer Lett 2024; 598:217109. [PMID: 39002692 DOI: 10.1016/j.canlet.2024.217109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
Gemcitabine serves as a first-line chemotherapeutic treatment for pancreatic cancer (PC), but it is prone to rapid drug resistance. Increasing the sensitivity of PC to gemcitabine has long been a focus of research. Fasting interventions may augment the effects of chemotherapy and present new options. SIRT7 is known to link metabolism with various cellular processes through post-translational modifications. We found upregulation of SIRT7 in PC cells is associated with poor prognosis and gemcitabine resistance. Cross-analysis of RNA-seq and ATAC-seq data suggested that GLUT3 might be a downstream target gene of SIRT7. Subsequent investigations demonstrated that SIRT7 directly interacts with the enhancer region of GLUT3 to desuccinylate H3K122. Our group's another study revealed that GLUT3 can transport gemcitabine in breast cancer cells. Here, we found GLUT3 KD reduces the sensitivity of PC cells to gemcitabine, and SIRT7 KD-associated gemcitabine-sensitizing could be reversed by GLUT3 KD. While fasting mimicking induced upregulation of SIRT7 expression in PC cells, knocking down SIRT7 enhanced sensitivity to gemcitabine through upregulating GLUT3 expression. We further confirmed the effect of SIRT7 deficiency on the sensitivity of gemcitabine under fasting conditions using a mouse xenograft model. In summary, our study demonstrates that SIRT7 can regulate GLUT3 expression by binding to its enhancer and altering H3K122 succinylation levels, thus affecting gemcitabine sensitivity in PC cells. Additionally, combining SIRT7 knockdown with fasting may improve the efficacy of gemcitabine. This unveils a novel mechanism by which SIRT7 influences gemcitabine sensitivity in PC and offer innovative strategies for clinical combination therapy with gemcitabine.
Collapse
Affiliation(s)
- Keyu Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing, 100191, China
| | - Tiane Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing, 100191, China
| | - Honglin Diao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing, 100191, China
| | - Qikai Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing, 100191, China
| | - Xiaojia Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing, 100191, China
| | - Zhihua Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing, 100191, China
| | - Mingyue Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing, 100191, China
| | - Zebin Mao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing, 100191, China.
| | - Yinmo Yang
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China.
| | - Wenhua Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
316
|
Heervä E, Väliaho V, Nurmi H, Lietzen E, Ålgars A, Kauhanen S. Outcomes After Multimodality Treatment of Pancreatic Cancer in an Unselected Single-Center Cohort. Cancer Manag Res 2024; 16:1065-1076. [PMID: 39220815 PMCID: PMC11363961 DOI: 10.2147/cmar.s465512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) remains a lethal and rarely resectable malignancy. Here we explore the outcomes of surgery, as compared to definitive radiotherapy (dRT) or systemic therapy only in PDAC. Methods Pancreatic surgery and radiotherapy in Southwest Finland have been centralized to Turku University Hospital. Previously validated population-based electronic health records database was searched for all unselected PDAC patients from the years 2009-2019. Main outcome was median overall survival (mOS). Demographics, pathology, surgery, and oncological treatment data were collected. Results We identified 1006 patients with PDAC, 49% male, median age 71 years and 77% presenting with metastatic disease. In total, 405 patients were treated; 92 resected, 26 dRT without resection and 287 systemic therapy only. mOS was 34.6 months for resected, 26.7 months for dRT, and 7.5 months for systemic therapy patients. Among the 88 patients with locally advanced inoperable PDAC, dRT was independently associated with longer mOS (26.7 months) as compared to systemic therapy only (mOS 10.6 months). Among the 287 patients treated with systemic therapy only, combination chemotherapy was independently associated with longer mOS (11.6 months) as compared to gemcitabine-monotherapy (6.8 months). In patients progressing to second-line systemic treatment after gemcitabine failure, mOS was the same (5.0 months) with single or combination regimens. Conclusion Surgery remains the only curative approach for PDAC. In locally advanced PDAC, dRT was associated with longer survival as compared to systemic therapy only. Concerning first-line systemic therapy, our results support the use of combination chemotherapy over single-agent therapy.
Collapse
Affiliation(s)
- Eetu Heervä
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Vesa Väliaho
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Heidi Nurmi
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Elina Lietzen
- Department of Digestive Surgery, Turku University Hospital and University of Turku, Turku, Finland
| | - Annika Ålgars
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Saila Kauhanen
- Department of Digestive Surgery, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
317
|
Peters GJ, Kathmann I, Giovannetti E, Smid K, Assaraf YG, Jansen G. The role of l-leucovorin uptake and metabolism in the modulation of 5-fluorouracil efficacy and antifolate toxicity. Front Pharmacol 2024; 15:1450418. [PMID: 39234107 PMCID: PMC11371747 DOI: 10.3389/fphar.2024.1450418] [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: 06/17/2024] [Accepted: 07/30/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND L-Leucovorin (l-LV; 5-formyltetrahydrofolate, folinic acid) is a precursor for 5,10-methylenetetrahydrofolate (5,10-CH2-THF), which is important for the potentiation of the antitumor activity of 5-fluorouracil (5FU). LV is also used to rescue antifolate toxicity. LV is commonly administered as a racemic mixture of its l-LV and d-LV stereoisomers. We compared dl-LV with l-LV and investigated whether d-LV would interfere with the activity of l-LV. METHODS Using radioactive substrates, we characterized the transport properties of l-LV and d-LV, and compared the efficacy of l-LV with d-LV to potentiate 5FU-mediated thymidylate synthase (TS) inhibition. Using proliferation assays, we investigated their potential to protect cancer cells from cytotoxicity of the antifolates methotrexate, pemetrexed (Alimta), raltitrexed (Tomudex) and pralatrexate (Folotyn). RESULTS l-LV displayed an 8-fold and 3.5-fold higher substrate affinity than d-LV for the reduced folate carrier (RFC/SLC19A1) and proton coupled folate transporter (PCFT/SLC46A1), respectively. In selected colon cancer cell lines, the greatest enhanced efficacy of 5FU was observed for l-LV (2-fold) followed by the racemic mixture, whereas d-LV was ineffective. The cytotoxicity of antifolates in lymphoma and various solid tumor cell lines could be protected very efficiently by l-LV but not by d-LV. This protective effect of l-LV was dependent on cellular RFC expression as corroborated in RFC/PCFT-knockout and RFC/PCFT-transfected cells. Assessment of TS activity in situ showed that TS inhibition by 5FU could be enhanced by l-LV and dl-LV and only partially by d-LV. However, protection from inhibition by various antifolates was solely achieved by l-LV and dl-LV. CONCLUSION In general l-LV acts similar to the dl-LV formulations, however disparate effects were observed when d-LV and l-LV were used in combination, conceivably by d-LV affecting (anti)folate transport and intracellular metabolism.
Collapse
Affiliation(s)
- Godefridus J. Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Ietje Kathmann
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Kees Smid
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Yehuda G. Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, The Technion-Israel Institute of Technology, Haifa, Israel
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
318
|
Grisendi G, Dall'Ora M, Casari G, Spattini G, Farshchian M, Melandri A, Masciale V, Lepore F, Banchelli F, Costantini RC, D'Esposito A, Chiavelli C, Spano C, Spallanzani A, Petrachi T, Veronesi E, Ferracin M, Roncarati R, Vinet J, Magistri P, Catellani B, Candini O, Marra C, Eccher A, Bonetti LR, Horwtiz EM, Di Benedetto F, Dominici M. Combining gemcitabine and MSC delivering soluble TRAIL to target pancreatic adenocarcinoma and its stroma. Cell Rep Med 2024; 5:101685. [PMID: 39168103 PMCID: PMC11384958 DOI: 10.1016/j.xcrm.2024.101685] [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: 05/31/2023] [Revised: 05/13/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) still has a poor response to therapies, partly due to their cancer-associated fibroblasts (CAFs). Here, we investigate the synergistic impact of a combinatory approach between a known chemotherapy agent, such as gemcitabine (GEM), and gene-modified human mesenchymal stromal/stem cells (MSCs) secreting the pro-apoptotic soluble (s)TRAIL (sTRAIL MSCs) on both PDAC cells and CAFs. The combo significantly impacts on PDAC survival in 2D and 3D models. In orthotopic xenograft models, GEM and sTRAIL MSCs induce tumor architecture shredding with a reduction of CK7- and CK8/18-positive cancer cells and the abrogation of spleen metastases. A cytotoxic effect on primary human CAFs is also observed along with an alteration of their transcriptome and a reduction of the related desmoplasia. Collectively, we demonstrate a promising therapeutic profile of combining GEM and sTRAIL MSCs to target both tumoral and stromal compartments in PDAC.
Collapse
Affiliation(s)
- Giulia Grisendi
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy.
| | | | - Giulia Casari
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Polytechnic University of Marche, Ancona
| | | | - Moein Farshchian
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Aurora Melandri
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Valentina Masciale
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Fabio Lepore
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Federico Banchelli
- Center of Statistic, Department of Medical and Surgical Sciences, UNIMORE, Modena, Italy
| | | | - Angela D'Esposito
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Chiara Chiavelli
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Carlotta Spano
- Department of Biomedical, Metabolic, and Neural Sciences, UNIMORE, Modena, Italy
| | | | | | | | - Manuela Ferracin
- Department of Medical and Surgical Sciences, University of Bologna, Bologna; IRCCS AOU di Bologna, Policlinico S. Orsola-Malpighi, Bologna
| | | | | | - Paolo Magistri
- Hepato-pancreato-biliary Surgery and Liver Transplantation Unit, UNIMORE, Modena, Italy
| | - Barbara Catellani
- Hepato-pancreato-biliary Surgery and Liver Transplantation Unit, UNIMORE, Modena, Italy
| | | | - Caterina Marra
- Division of Plastic Surgery, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | | | | | - Edwin M Horwtiz
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Fabrizio Di Benedetto
- Hepato-pancreato-biliary Surgery and Liver Transplantation Unit, UNIMORE, Modena, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy; Division of Oncology, University-Hospital of Modena, Modena, Italy; Division of Medical Oncology, Residency School of Medical Oncology, Program in Cellular Therapy and Immuno-oncology, Laboratory of Cellular Therapy, University Hospital of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
319
|
Cortiana V, Vallabhaneni H, Gambill J, Nadar S, Itodo K, Park CH, Leyfman Y. Advancing Pancreatic Cancer Surgical Treatments and Proposal of New Approaches. Cancers (Basel) 2024; 16:2848. [PMID: 39199619 PMCID: PMC11352325 DOI: 10.3390/cancers16162848] [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: 06/26/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Pancreatic cancer is a significant challenge in oncology due to its aggressive nature and complex management, leading to high mortality rates and a dismally low 5-year survival rate. Approximately 85% of cases manifest as adenocarcinoma, while endocrine tumors constitute less than 5%. Borderline resectable and locally advanced pancreatic cancers are particularly difficult to treat due to vascular involvement, which complicates complete resections and increases morbidity. Various therapeutic modalities aim to overcome these challenges and improve patient outcomes. Traditionally, upfront surgery was the standard for resectable tumors, with multimodal chemotherapy being central to treatment. Understanding surgical anatomy is pivotal in enhancing surgical outcomes and patient survival. Resectability challenges are several when seeking to achieve R0 resections, particularly for borderline resectable tumors. Various classification systems-the MD Anderson criteria, the NCCN criteria, the AHPA/SSAT/SSO consensus statement, and the Alliance definition-assess tumor involvement with major blood vessels, with the first of these systems being broadly accepted. Vascular staging integration is also important, with the Ishikawa staging system using preoperative imaging to assess venous involvement. Furthermore, neoadjuvant therapy enhances treatment effectiveness by addressing micro-metastatic disease early, increasing R0 resection chances, and downstaging tumors for optimal surgery. Insights from the Fox Chase Cancer Center's neoadjuvant treatment approach highlight the importance of a multidisciplinary strategy when advancing therapy and improving patient prognosis. This commentary, inspired by Dr. Sanjay S. Reddy's Keynote Conference during MedNews week, highlights current advancements and ongoing challenges in the treatment of pancreatic cancer, emphasizing the need for a comprehensive, multidisciplinary approach to improve outcomes.
Collapse
Affiliation(s)
- Viviana Cortiana
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | | | | | - Soumiya Nadar
- Tbilisi State Medical University, 0186 Tbilisi, Georgia
| | - Kennedy Itodo
- Nigerian Institute for Trypanosomiasis Research Jos, Kaduna PMB 2077, Nigeria
| | | | - Yan Leyfman
- Icahn School of Medicine at Mount Sinai South Nassau, Oceanside, NY 11572, USA;
| |
Collapse
|
320
|
van Eijck CWF, Haddaoui HE, Kucukcelebi S, Vadgama D, Fellah A, Mustafa DAM, Aerts JGJV, van Eijck CHJ, Willemsen M. Rintatolimod in Advanced Pancreatic Cancer Enhances Antitumor Immunity through Dendritic Cell-Mediated T-Cell Responses. Clin Cancer Res 2024; 30:3447-3458. [PMID: 38488815 DOI: 10.1158/1078-0432.ccr-23-4085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/05/2024] [Accepted: 03/13/2024] [Indexed: 08/16/2024]
Abstract
PURPOSE Amid the need for new approaches to improve survival in pancreatic ductal adenocarcinoma (PDAC), immune-based therapies have garnered interest. Rintatolimod, a Toll-like receptor 3 (TLR-3) agonist, is a potential candidate due to its dual impact on restraining PDAC cell functions and boosting the antitumor immune response. This study investigates the effect of TLR-3 activation through rintatolimod on the peripheral immune landscape of patients with advanced PDAC. EXPERIMENTAL DESIGN Paired blood samples of 30 patients with advanced PDAC, collected at baseline and after 12 rintatolimod intravenous infusions, underwent comprehensive transcriptomic NanoString and proteomic flow cytometry profiling. The impact of rintatolimod and immunologic factors on survival outcomes was assessed through univariate Cox proportional hazards models. RESULTS Rintatolimod treatment enhances peripheral immune activity at the transcriptomic and proteomic levels, particularly involving type 1 conventional dendritic cells (cDC1) and T cells. Post-rintatolimod, the increased peripheral abundance of BTLA+ XCR1+ cDC1s and CD4+SELL+ T cells correlated with improved clinical outcomes. Patients with stable disease exhibited pronouncedDCand T-cell activation gene overexpression. Notably, the expression of immune checkpoints PD-L1 and PD-L2 decreased post-rintatolimod across all patients. However, those with progressive disease showed increased expression of genes encoding IDO1 and PD-1. CONCLUSIONS This study presents compelling evidence of the immune-stimulatory properties linked to TLR-3 activation through rintatolimod. Rintatolimod may break immunologic tolerance by enhancing antitumor immunity through DC-mediated Th-cell responses. Furthermore, our findings lay the groundwork for investigating the potential synergy between TLR-3 activation and immune checkpoint inhibitor therapy to improve therapeutic outcomes. See related commentary by Martínez-Riaño et al., p. 3355.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Hassana El Haddaoui
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Songul Kucukcelebi
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Disha Vadgama
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Amine Fellah
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Dana A M Mustafa
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Department of Clinical Bioinformatics, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Joachim G J V Aerts
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Marcella Willemsen
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
321
|
Haller SD, Essani K. Oncolytic Tanapoxvirus Variants Expressing mIL-2 and mCCL-2 Regress Human Pancreatic Cancer Xenografts in Nude Mice. Biomedicines 2024; 12:1834. [PMID: 39200298 PMCID: PMC11351728 DOI: 10.3390/biomedicines12081834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/01/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fifth leading cause of cancer-related death and presents the lowest 5-year survival rate of any form of cancer in the US. Only 20% of PDAC patients are suitable for surgical resection and adjuvant chemotherapy, which remains the only curative treatment. Chemotherapeutic and gene therapy treatments are associated with adverse effects and lack specificity/efficacy. In this study, we assess the oncolytic potential of immuno-oncolytic tanapoxvirus (TPV) recombinants expressing mouse monocyte chemoattractant protein (mMCP-1 or mCCL2) and mouse interleukin (mIL)-2 in human pancreatic BxPc-3 cells using immunocompromised and CD-3+ T-cell-reconstituted mice. Intratumoral treatment with TPV/∆66R/mCCL2 and TPV/∆66R/mIL-2 resulted in a regression in BxPc-3 xenograft volume compared to control in immunocompromised mice; mCCL-2 expressing TPV OV resulted in a significant difference from control at p < 0.05. Histological analysis of immunocompromised mice treated with TPV/∆66R/mCCL2 or TPV/∆66R/mIL-2 demonstrated multiple biomarkers indicative of increased severity of chronic, active inflammation compared to controls. In conclusion, TPV recombinants expressing mCCL2 and mIL-2 demonstrated a therapeutic effect via regression in BxPc-3 tumor xenografts. Considering the enhanced oncolytic potency of TPV recombinants demonstrated against PDAC in this study, further investigation as an alternative or combination treatment option for human PDAC may be warranted.
Collapse
Affiliation(s)
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008-5410, USA;
| |
Collapse
|
322
|
Kawamoto Y, Yamai T, Ikezawa K, Seiki Y, Watsuji K, Hirao T, Urabe M, Kai Y, Takada R, Mukai K, Nakabori T, Uehara H, Inoue T, Fujisawa F, Ohkawa K. Clinical significance of germline breast cancer susceptibility gene (gBRCA) testing and olaparib as maintenance therapy for patients with pancreatic cancer. BMC Cancer 2024; 24:1000. [PMID: 39134950 PMCID: PMC11321060 DOI: 10.1186/s12885-024-12722-8] [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: 02/10/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Germline breast cancer susceptibility gene (gBRCA) mutation in patients with pancreatic cancer (PC) is not common in clinical practice. Therefore, factors that efficiently show gBRCA mutations and the real-world outcomes of olaparib maintenance therapy have not been fully established. In the present study, we clarified the indicators for the effective detection of gBRCA mutation and the efficacy and safety of olaparib as maintenance therapy. METHODS We retrospectively analyzed 84 patients with PC who underwent gBRCA testing (BRACAnalysis, Myriad Genetics, Salt Lake City, UT, USA) at our institute between January 2021 and March 2022. For each patient, clinical data were extracted from medical records. RESULTS The median patient age was 64 y (29-85 y), and 41 patients (48.8%) were male. The gBRCA mutations were identified in 10 (11.9%) patients; two patients had BRCA1 mutation and eight had BRCA2 mutation. All patients with gBRCA mutation had a family history of any cancer, and eight of them had a family history of Hereditary Breast and Ovarian Cancer syndrome (HBOC)-related cancer. The gBRCA mutation rate was higher for patients with PC with a family history of HBOC-related cancer compared to that in patients with PC having a family history of other cancers and no family history of cancer (22.9% vs. 4.1%; P = 0.014). In our study, eight out of 10 patients with gBRCA-positive PC received olaparib after platinum-based chemotherapy. The best responses to platinum-based chemotherapy included a complete response in one patient (12.5%) and a partial response in seven patients (87.5%). The median duration of treatment with platinum-based chemotherapy plus olaparib was 17.5 months (8-87 months), and the duration of treatment with olaparib maintenance therapy was 11 months (1-30 months). During olaparib maintenance therapy, three patients showed no disease progression. One of these three patients underwent conversion surgery after receiving olaparib for 12 months. CONCLUSIONS The gBRCA testing should be considered proactively, especially in patients with PC with a family history of HBOC-related cancer.
Collapse
Affiliation(s)
- Yasuharu Kawamoto
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Takuo Yamai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
- Department of Gastroenterology, Osaka General Medical Center, Osaka, Japan
| | - Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan.
| | - Yusuke Seiki
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Ko Watsuji
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Takeru Hirao
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Makiko Urabe
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Yugo Kai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Ryoji Takada
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Kaori Mukai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Tasuku Nakabori
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Hiroyuki Uehara
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| | - Tazuko Inoue
- Department of Genetic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Fumie Fujisawa
- Department of Genetic Oncology, Osaka International Cancer Institute, Osaka, Japan
- Department of Medical Oncology, Shiga General Hospital, Shiga, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chou-ku, Osaka, 541-8567, Japan
| |
Collapse
|
323
|
Brugiapaglia S, Bulfamante S, Curcio C, Arigoni M, Calogero R, Bonello L, Genuardi E, Spadi R, Satolli MA, Campra D, Giordano D, Cappello P, Cordero F, Novelli F. In pancreatic cancer patients, chemotherapy reshapes the gene expression profile and antigen receptor repertoire of T lymphocytes and enhances their effector response to tumor-associated antigens. Front Immunol 2024; 15:1427424. [PMID: 39176093 PMCID: PMC11339620 DOI: 10.3389/fimmu.2024.1427424] [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: 05/03/2024] [Accepted: 07/15/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction Pancreatic Ductal Adenocarcinoma (PDA) is one of the most aggressive malignancies with a 5-year survival rate of 13%. Less than 20% of patients have a resectable tumor at diagnosis due to the lack of distinctive symptoms and reliable biomarkers. PDA is resistant to chemotherapy (CT) and understanding how to gain an anti-tumor effector response following stimulation is, therefore, critical for setting up an effective immunotherapy. Methods Proliferation, and cytokine release and TCRB repertoire of from PDA patient peripheral T lymphocytes, before and after CT, were analyzed in vitro in response to four tumor-associated antigens (TAA), namely ENO1, FUBP1, GAPDH and K2C8. Transcriptional state of PDA patient PBMC was investigated using RNA-Seq before and after CT. Results CT increased the number of TAA recognized by T lymphocytes, which positively correlated with patient survival, and high IFN-γ production TAA-induced responses were significantly increased after CT. We found that some ENO1-stimulated T cell clonotypes from CT-treated patients were expanded or de-novo induced, and that some clonotypes were reduced or even disappeared after CT. Patients that showed a higher number of effector responses to TAA (high IFN-γ/IL-10 ratio) after CT expressed increased fatty acid-related transcriptional signature. Conversely, patients that showed a higher number of regulatory responses to TAA (low IFN-γ/IL-10 ratio) after CT significantly expressed an increased IRAK1/IL1R axis-related transcriptional signature. Conclusion These data suggest that the expression of fatty acid or IRAK1/IL1Rrelated genes predicts T lymphocyte effector or regulatory responses to TAA in patients that undergo CT. These findings are a springboard to set up precision immunotherapies in PDA based on the TAA vaccination in combination with CT.
Collapse
MESH Headings
- Humans
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/therapy
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Male
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Carcinoma, Pancreatic Ductal/therapy
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/drug therapy
- Female
- Transcriptome
- Aged
- Middle Aged
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Gene Expression Regulation, Neoplastic
- Gene Expression Profiling
- Phosphopyruvate Hydratase/genetics
- Phosphopyruvate Hydratase/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Silvia Brugiapaglia
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
| | - Sara Bulfamante
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Claudia Curcio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Raffaele Calogero
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Lisa Bonello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Elisa Genuardi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Rosella Spadi
- Centro Oncologico Ematologico Subalpino, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, Turin, Italy
| | - Maria Antonietta Satolli
- Centro Oncologico Ematologico Subalpino, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, Turin, Italy
| | - Donata Campra
- Struttura Complessa (SC) Chirurgia generale d’urgenza e pronto soccorso, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, Turin, Italy
| | - Daniele Giordano
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
| | | | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
| |
Collapse
|
324
|
Tosca EM, Ronchi D, Rocchetti M, Magni P. Predicting Tumor Volume Doubling Time and Progression-Free Survival in Untreated Patients from Patient-Derived-Xenograft (PDX) Models: A Translational Model-Based Approach. AAPS J 2024; 26:92. [PMID: 39117850 DOI: 10.1208/s12248-024-00960-4] [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: 03/13/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Tumor volume doubling time (TVDT) has been shown to be a potential surrogate marker of biological tumor activity. However, its availability in clinics is strongly limited due to ethical and practical reasons, as its assessment requires at least two subsequent tumor volume measurements in untreated patients. Here, a translational modeling framework to predict TVDT distributions in untreated cancer patient populations from tumor growth data in patient-derived xenograft (PDX) mice is proposed. Eleven solid cancer types were considered. For each of them, a set of tumor growth studies in PDX mice was selected and analyzed through a mathematical model to characterize the distribution of the exponential tumor growth rate in mice. Then, assuming an exponential growth of the tumor mass in humans, the growth rates were scaled from PDX mice to humans through an allometric scaling approach and used to predict TVDTs in untreated patients. A very good agreement was found between model predicted and clinically observed TVDTs, with 91% of the predicted TVDT medians fell within 1.5-fold of observations. Further, exploiting the intrinsic relationship between tumor growth dynamics and progression free survival (PFS), the exponential growth rates in humans were used to generate the expected PFS curves in absence of anticancer treatment. Predicted curves were extremely close to published PFS data from studies involving patient cohorts treated with supportive care or low effective therapies. The proposed approach shows promise as a potential tool to increase knowledge about TVDT in humans without the need of directly measuring tumor dimensions in untreated patients, and to predict PFS curves in untreated patients, that could fill the absence of placebo-controlled arms against which to compare treaded arms during clinical trials. However, further validation and refinement are needed to fully assess its effectiveness in this regard.
Collapse
Affiliation(s)
- E M Tosca
- Dipartimento Di Ingegneria Industriale E Dell'Informazione, Università Degli Studi Di Pavia, 27100, Pavia, Italy
| | - D Ronchi
- Dipartimento Di Ingegneria Industriale E Dell'Informazione, Università Degli Studi Di Pavia, 27100, Pavia, Italy
| | | | - P Magni
- Dipartimento Di Ingegneria Industriale E Dell'Informazione, Università Degli Studi Di Pavia, 27100, Pavia, Italy.
| |
Collapse
|
325
|
Zhou Y, Ma Y, Sheng J, Ma Y, Ding J, Zhou W. Breaking Down Barriers in Drug Delivery by Stromal Remodeling Approaches in Pancreatic Cancer. Mol Pharm 2024; 21:3764-3776. [PMID: 39049481 DOI: 10.1021/acs.molpharmaceut.4c00329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Pancreatic cancer remains a formidable challenge in oncology due to its aggressive nature and limited treatment options. The dense stroma surrounding pancreatic tumors not only provides structural support but also presents a formidable barrier to effective therapy, hindering drug penetration and immune cell infiltration. This review delves into the intricate interplay between stromal components and cancer cells, highlighting their impact on treatment resistance and prognosis. Strategies for stromal remodeling, including modulation of cancer-associated fibroblasts (CAFs), pancreatic stellate cells (PSCs) activation states, and targeting extracellular matrix (ECM) components, are examined for their potential to enhance drug penetration and improve therapeutic efficacy. Integration of stromal remodeling with conventional therapies, such as chemotherapy and immunotherapy, is discussed along with the emerging field of intelligent nanosystems for targeted drug delivery. This comprehensive overview underscores the importance of stromal remodeling in pancreatic cancer treatment and offers insights into promising avenues for future research and clinical translation.
Collapse
Affiliation(s)
- Ying Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Yunxiao Ma
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Jianwei Sheng
- China Quality Mark Certification (Shandong) Co., LTD, Jinan, Shandong 250100, China
| | - Yiran Ma
- Hunan Bainianyiren Chinese Traditional Medical Institute Co., LTD, Changsha, Hunan 410221, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Changsha, Hunan 410008, China
| |
Collapse
|
326
|
Hatoum H, Rosemurgy A, Bastidas JA, Zervos E, Muscarella P, Edil BH, Cynamon J, Johnson DT, Thomas C, Swinson BM, Nordgren A, Vitulli P, Nutting C, Gipson M, Tsobanoudis A, Agah R. Treatment of locally advanced pancreatic cancer using localized trans-arterial micro perfusion of gemcitabine: combined analysis of RR1 and RR2. Oncologist 2024; 29:690-698. [PMID: 39049803 PMCID: PMC11299923 DOI: 10.1093/oncolo/oyae178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Locally advanced pancreatic cancer (LAPC) comprises 40% of pancreatic cancer diagnoses and has a relatively poor prognosis. Trans-arterial micro perfusion (TAMP)-mediated chemotherapy delivery to the primary tumor is a novel approach worthy of investigation. The RR1 (dose escalation) and RR2 (observational) studies examined the safety and preliminary efficacy of TAMP-delivered gemcitabine for LAPC. PATIENTS AND METHODS RR1 and RR2 data were pooled. Both studies enrolled patients with LAPC with histologically confirmed adenocarcinoma. Participant data, including age, sex, race, stage, previous treatments, toxicity, disease progression, and death, were collected. Median number of cycles and average treatment dosage were calculated. Overall survival (OS) was determined for the whole group and separately for patients who received and did not receive previous treatments. Aims of the analysis were to assess procedure safety, OS, and evaluate factors associated with OS. RESULTS The median age of the 43 patients enrolled in RR1 and RR2 was 72 years (range, 51-88 years). Median OS for the 35 eligible patients with stage III disease was 12.6 months (95% CI, 2.1-54.2 months). Previous chemoradiation was associated with significantly longer OS [27.1 months (95% CI, 8.4-40.6 months)] compared to previous systemic chemotherapy [14.6 months (95% CI, 6.4-54.2 months)] or no prior treatment [7.0 months (95% CI, 2.1-35.4 months)] (P < .001). The most common adverse events were GI related (abdominal pain, emesis, and vomiting); the most common grade 3 toxicity was sepsis. CONCLUSION Study results indicate that TAMP-mediated gemcitabine delivery in patients with LAPC is potentially safe, feasible, and provides potential clinical benefits. CLINICAL TRIAL REGISTRATION NCT02237157 (RR1) and NCT02591082 (RR2).
Collapse
Affiliation(s)
- Hassan Hatoum
- Department of Internal Medicine Hematology-Oncology Section, University of Oklahoma College of Medicine, Oklahoma City, OK, United States
| | | | | | - Emmanuel Zervos
- Division of Surgical Oncology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Peter Muscarella
- Montefiore Medical Center, Bronx, NY, United States
- Niagara Falls Memorial Medical Center, Niagara Falls, NY, United States
| | - Barish H Edil
- Department of Surgery, University of Oklahoma College of Medicine, Oklahoma City, OK, United States
| | | | - D Thor Johnson
- University of Colorado Hospital, Aurora, CO, United States
- Sarah Cannon, Nashville, TN, United States
| | | | | | - Aaron Nordgren
- Fawcett Memorial Hospital, Port Charlotte, FL, United States
| | - Paul Vitulli
- Florida Hospital, Tampa, Tampa, FL, United States
- Duval Vascular Center, Jacksonville, FL, United States
| | - Charles Nutting
- Endovascular Consultants of Colorado, Lone Tree, CO, United States
| | | | | | - Ramtin Agah
- RenovoRx, Inc., Los Altos, CA, United States
| |
Collapse
|
327
|
Girolimetti G, Balena B, Cordella P, Verri T, Eusebi LH, Bozzetti MP, Bucci C, Guerra F. Characterization of Chemoresistance in Pancreatic Cancer: A Look at MDR-1 Polymorphisms and Expression in Cancer Cells and Patients. Int J Mol Sci 2024; 25:8515. [PMID: 39126083 PMCID: PMC11312866 DOI: 10.3390/ijms25158515] [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: 07/03/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Pancreatic malignancy is the fourth cause of cancer-related death in Western countries and is predicted to become the second leading cause of cancer-related mortality by 2030. The standard therapies (FOLFIRINOX and gemcitabine with nab-paclitaxel) are not resolutive because this type of cancer is also characterized by a high chemoresistance, due in part to the activity of the ATP Binding Cassette (ABC) pumps accounting for the reduction in the intracellular concentration of the drugs. In this work, we analyze the occurrence of single-nucleotide polymorphisms (SNPs) in the MDR-1 gene, in different pancreatic cancer cell lines, and in tissues from pancreatic cancer patients by DNA sequencing, as well as the expression levels of MDR-1 mRNA and protein, by qRT-PCR and Western Blot analysis. We found that gemcitabine-resistant cells, in conjunction with homozygosis of analyzed SNPs, showed high MDR-1 basal levels with further increases after gemcitabine treatment. Nevertheless, we did not observe in the human PDAC samples a correlation between the level of MDR-1 mRNA and protein expression and SNPs. Preliminary, we conclude that in our small cohort, these SNPs cannot be used as molecular markers for predicting the levels of MDR-1 mRNA/protein levels and drug responses in patients with PDAC.
Collapse
Affiliation(s)
- Giulia Girolimetti
- Department of Experimental Medicine (DiMeS), University of Salento, Via Provinciale Lecce-Monteroni n.165, 73100 Lecce, Italy; (G.G.); (P.C.)
| | - Barbara Balena
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni n.165, 73100 Lecce, Italy; (B.B.); (T.V.); (M.P.B.)
| | - Paola Cordella
- Department of Experimental Medicine (DiMeS), University of Salento, Via Provinciale Lecce-Monteroni n.165, 73100 Lecce, Italy; (G.G.); (P.C.)
| | - Tiziano Verri
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni n.165, 73100 Lecce, Italy; (B.B.); (T.V.); (M.P.B.)
| | - Leonardo Henry Eusebi
- Gastroenterology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Maria Pia Bozzetti
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni n.165, 73100 Lecce, Italy; (B.B.); (T.V.); (M.P.B.)
| | - Cecilia Bucci
- Department of Experimental Medicine (DiMeS), University of Salento, Via Provinciale Lecce-Monteroni n.165, 73100 Lecce, Italy; (G.G.); (P.C.)
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni n.165, 73100 Lecce, Italy; (B.B.); (T.V.); (M.P.B.)
| |
Collapse
|
328
|
Huang C, Cheng CS, Shen Y, Chen H, Lin J, Hua Y, Feng L, Wu C, Wang P, Chen Z, Meng Z. Digital subtraction angiography-guided pancreatic arterial infusion of GEMOX chemotherapy in advanced pancreatic adenocarcinoma: a phase II, open-label, randomized controlled trial comparing with intravenous chemotherapy. BMC Cancer 2024; 24:941. [PMID: 39095759 PMCID: PMC11295591 DOI: 10.1186/s12885-024-12695-8] [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: 09/07/2023] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Advanced pancreatic adenocarcinoma lacks effective treatment options, and systemic gemcitabine-based chemotherapy offers only marginal survival benefits at the cost of significant toxicities and adverse events. New therapeutic options with better drug availability are warranted. This study aims to evaluate the safety and efficacy of digital subtraction angiography (DSA)-guided pancreatic arterial infusion (PAI) versus intravenous chemotherapy (IVC) using the gemcitabine and oxaliplatin (GEMOX) regimen in unresectable locally advanced or metastatic pancreatic cancer (PC) patients. MATERIALS AND METHODS This study prospectively enrolled 51 eligible treatment-naive patients with unresectable PC to receive GEMOX treatment via PAI or IVC (1:1 ratio randomization) from December 2015 to December 2019. Cycles were repeated monthly, and each process consisted of two treatments administered bi-weekly. Overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), 1-year survival, 6-month survival, tumor-site subgroup survival, and incidences of adverse events were compared. RESULTS The median OS of the PAI and IVC groups were 9.93 months and 10.07 months, respectively (p = 0.3049). The median PFS of the PAI and IVC groups were 5.07 months and 4.23 months (p = 0.1088). No significant differences were found in the ORR (11.54% vs. 4%, p = 0.6312), DCR (53.85% vs. 44%, p = 0.482), and 1-year OS rate (44% vs. 20.92%, p = 0.27) in PAI and IVC groups. The 6-month OS rate was significantly higher in the PAI group (100%) than in the IVC group (83.67%) (p = 0.0173). The median OS of patients in PAI group with pancreatic head and neck tumors were significantly higher than those of body and tail tumors (12.867 months vs. 9 months, p = 0.0214). The incidences of hematologic disorders, liver function disorders, and digestive disorders in the IVC group were higher than in the PAI group (p < 0.05). CONCLUSION GEMOX PAI therapy presented a higher 6-month OS rate and fewer adverse events than IVC in advanced pancreatic adenocarcinoma patients. Those with pancreatic head and neck tumors may yield a superior treatment outcome from PAI treatment. TRIAL REGISTRATION NUMBER NCT02635971. DATE OF REGISTRATION 21/12/2015.
Collapse
Affiliation(s)
- Changjing Huang
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Chien-Shan Cheng
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yehua Shen
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Hao Chen
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Junhua Lin
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yongqiang Hua
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lanyun Feng
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Caijun Wu
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Peng Wang
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhen Chen
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhiqiang Meng
- Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
329
|
Paredes de la Fuente R, Sucre S, Ponce C, Rattani AAA, Peters MLB. Somatic Mutation Profile as a Predictor of Treatment Response and Survival in Unresectable Pancreatic Ductal Adenocarcinoma Treated with FOLFIRINOX and Gemcitabine Nab-Paclitaxel. Cancers (Basel) 2024; 16:2734. [PMID: 39123462 PMCID: PMC11312283 DOI: 10.3390/cancers16152734] [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: 07/11/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
(1) Background: Pancreatic ductal adenocarcinoma (PDAC) has low survival rates despite treatment advancements. Aim: This study aims to show how molecular profiling could possibly guide personalized treatment strategies, which may help improve survival outcomes in patients with PDAC. (2) Materials and Methods: A retrospective analysis of 142 PDAC patients from a single academic center was conducted. Patients underwent chemotherapy and next-generation sequencing for molecular profiling. Key oncogenic pathways were identified using the Reactome pathway database. Survival analysis was performed using Kaplan-Meier curves and Cox Proportional Hazards Regression. (3) Results: Patients mainly received FOLFIRINOX (n = 62) or gemcitabine nab-paclitaxel (n = 62) as initial chemotherapy. The median OS was 13.6 months. Longer median OS was noted in patients with NOTCH (15 vs. 12.3 months, p = 0.007) and KIT pathway mutations (21.3 vs. 12.12 months, p = 0.04). Combinatorial pathway analysis indicated potential synergistic effects on survival. In the PFS, PI3K pathway (6.6 vs. 5.7 months, p = 0.03) and KIT pathway (10.3 vs. 6.2 months, p = 0.03) mutations correlated with improved PFS within the gemcitabine nab-paclitaxel subgroup. (4) Conclusions: Molecular profiling could play a role in PDAC for predicting outcomes and responses to therapies like FOLFIRINOX and gemcitabine nab-paclitaxel. Integrating genomic data into clinical decision-making can benefit PDAC treatment, though further validation is needed to fully utilize precision oncology in PDAC management.
Collapse
Affiliation(s)
| | - Santiago Sucre
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA (A.A.A.R.)
| | - Cristina Ponce
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA (A.A.A.R.)
| | - Ahmed Anwer Ali Rattani
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA (A.A.A.R.)
| | - Mary Linton B. Peters
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA (A.A.A.R.)
| |
Collapse
|
330
|
Suzuki S, Nagakawa Y, Miyamoto R, Osakabe H, Kiya Y, Yamaguchi H, Nagao T, Einama T, Ao T, Shimoda M. Prognostic factors based on histological categorization of desmoplastic reactions in pancreatic cancer. World J Surg 2024; 48:1973-1980. [PMID: 38943046 DOI: 10.1002/wjs.12269] [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: 06/16/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND/PURPOSE In colorectal cancer, the morphological categorization of fibrotic cancer stroma in the invasive frontal zone of the primary tumor is well reflected in the prognosis. Conversely, the histological characteristics of pancreatic cancer (PC) reveal fibrotic hyperplasia of stroma known as desmoplasia; however, its characterization is unknown. Therefore, this study aimed to evaluate the prognostic factors according to the histological categorization of desmoplastic reactions in PC. METHODS We retrospectively enrolled 167 patients who underwent curative resection for PC. The desmoplastic pattern was histologically classified as mature, intermediate, or immature. Clinicopathological features were evaluated, and disease-free and overall survival (OS) were analyzed in the three groups. Prognostic factors were assessed using univariate and multivariate analyses. RESULTS In total, 19 mature, 87 intermediate, and 61 immature desmoplastic patterns were evaluated. Jaundice decompression, white blood cell count, and platelet/lymphocyte ratio were significantly different among the groups. The mature group had a better disease-free survival (DFS) prognosis than the other two groups; however, OS did not differ between the groups. Desmoplastic patterns showed significant differences between the three groups for DFS. CONCLUSIONS Desmoplastic patterns are a prognostic factor of DFS for PC, with mature desmoplastic reactions associated with good prognosis. Thus, they may aid in individualized therapeutic approaches in patients with PC.
Collapse
Affiliation(s)
- Shuji Suzuki
- Department of Gastroenterological Surgery, Ibaraki Medical Center, Tokyo Medical University, Inashikigun, Ibaraki, Japan
| | - Yuichi Nagakawa
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo, Japan
| | - Ryoichi Miyamoto
- Department of Gastroenterological Surgery, Ibaraki Medical Center, Tokyo Medical University, Inashikigun, Ibaraki, Japan
| | - Hiroaki Osakabe
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo, Japan
| | - Yoshitaka Kiya
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi Yamaguchi
- Department of Diagnostic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Toshitaka Nagao
- Department of Diagnostic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Takahiro Einama
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Tadakazu Ao
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Mitsugi Shimoda
- Department of Gastroenterological Surgery, Ibaraki Medical Center, Tokyo Medical University, Inashikigun, Ibaraki, Japan
| |
Collapse
|
331
|
Nevala-Plagemann C, Garrido-Laguna I. NALIRIFOX for metastatic pancreatic adenocarcinoma: hope or hype? Nat Rev Clin Oncol 2024; 21:567-568. [PMID: 38664532 DOI: 10.1038/s41571-024-00896-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Affiliation(s)
| | - Ignacio Garrido-Laguna
- Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
332
|
Gordon JW, Chen HY, Nickles T, Lee PM, Bok R, Ohliger MA, Okamoto K, Ko AH, Larson PEZ, Wang ZJ. Hyperpolarized 13C Metabolic MRI of Patients with Pancreatic Ductal Adenocarcinoma. J Magn Reson Imaging 2024; 60:741-749. [PMID: 38041836 PMCID: PMC11144260 DOI: 10.1002/jmri.29162] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 12/04/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDA) is the third leading cause of cancer-related death in the United States. However, early response assessment using the current approach of measuring changes in tumor size on computed tomography (CT) or MRI is challenging. PURPOSE To investigate the feasibility of hyperpolarized (HP) [1-13C]pyruvate MRI to quantify metabolism in the normal appearing pancreas and PDA, and to assess changes in PDA metabolism following systemic chemotherapy. STUDY TYPE Prospective. SUBJECTS Six patients (65.0 ± 7.6 years, 2 females) with locally advanced or metastatic PDA enrolled prior to starting a new line of systemic chemotherapy. FIELD STRENGTH/SEQUENCE 3-T, T1-weighted gradient echo, metabolite-selective 13C echoplanar imaging. ASSESSMENT Time-resolved HP [1-13C]pyruvate data were acquired before (N = 6) and 4-weeks after (N = 3) treatment initiation. Pyruvate metabolism, as quantified by pharmacokinetic modeling and metabolite area-under-the-curve ratios, was assessed in manually segmented PDA and normal appearing pancreas ROIs (N = 5). The change in tumor metabolism before and 4-weeks after treatment initiation was assessed in primary PDA (N = 2) and liver metastases (N = 1), and was compared to objective tumor response defined by response evaluation criteria in solid tumors (RECIST) on subsequent CTs. STATISTICAL TESTS Descriptive tests (mean ± standard deviation), model fit error for pharmacokinetic rate constants. RESULTS Primary PDA showed reduced alanine-to-lactate ratios when compared to normal pancreas, due to increased lactate-to-pyruvate or reduced alanine-to-pyruvate ratios. Of the three patients who received HP [1-13C]pyruvate MRI before and 4-weeks after treatment initiation, one patient had a primary tumor with early metabolic response (increase in alanine-to-lactate) and subsequent partial response according to RECIST, one patient had a primary tumor with relatively stable metabolism and subsequent stable disease by RECIST, and one patient had metastatic PDA with increase in lactate-to-pyruvate of the liver metastases and corresponding progressive disease according to RECIST. DATA CONCLUSION Altered pyruvate metabolism with increased lactate or reduced alanine was observed in the primary tumor. Early metabolic response assessed at 4-weeks after treatment initiation correlated with subsequent objective tumor response assessed using RECIST. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jeremy W Gordon
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Hsin-Yu Chen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Tanner Nickles
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Philip M Lee
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Robert Bok
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Michael A Ohliger
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Kimberly Okamoto
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Andrew H Ko
- Department of Medicine, University of California, San Francisco, California, USA
| | - Peder E Z Larson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, California, USA
| | - Zhen J Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, California, USA
| |
Collapse
|
333
|
Barros AG, Mansinho H, Couto N, Teixeira MR, Tonin FS, Francisco R, Duarte-Ramos F. The Initial Journey of Patients with Metastatic Pancreatic Cancer (PaCTO Project): A Nationwide Survey among Portuguese Specialist Physicians. GE PORTUGUESE JOURNAL OF GASTROENTEROLOGY 2024; 31:262-272. [PMID: 39114325 PMCID: PMC11305690 DOI: 10.1159/000533178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2024]
Abstract
Introduction We aimed to characterize the initial healthcare journey of metastatic pancreatic ductal adenocarcinoma (mPDAC) patients in Portugal, including healthcare provision and factors affecting therapeutic decisions, namely BRCA mutations testing. Methods This is a descriptive cross-sectional, web-based survey using a convenience sampling approach. Portuguese oncologists and pathologists that routinely work with mPDAC patients from the different geographical regions and settings were invited to participate in the study via email (December 2020). Descriptive statistical analyses were performed, with categorical variables reported as absolute and relative frequencies, and continuous variables with non-normal distribution as median and interquartile range (IQR) (Stata v.15.0). Results Seventy physicians participated in the study (43 oncologists, 27 pathologists). According to the responses, a median of 28 patients per center (IQR 12-70) was diagnosed with PDAC in the previous year; 22 of them referring (IQR 8-70) to mPDAC. The pointed median time from patients' first hospital admission until disease diagnosis/staging is between 2 and 4 weeks. Endoscopic ultrasound with fine-needle biopsy is available in most hospitals (86%). Around 50% of physicians request BRCA testing; the assessment of additional biomarkers besides BRCA is requested by 40% of professionals. Half of them stated that BRCA testing should be requested earlier-upon histological diagnosis, especially because the median time for results is of 4.0 weeks (IQR 4-8). PARP inhibitors such as olaparib, when available, would be the therapy of choice for most oncologists (71%) if no disease' progression occurs after 4 months. Treatments' selection is usually grounded on clinical criteria (e.g., performance status, liver function). Around 45% of patients use FOLFIRINOX/mFOLFIRINOX as the first-line therapy. Gemcitabine + nab-paclitaxel is used by 35% of patients as the second-line therapy. Conclusions Physicians in Portugal support the increasing role of patient-tailored treatments in mPDAC, whose selection should be grounded on tumoral subtyping and molecular profiling. Further efforts to develop multidisciplinary teams, standardized clinical practice, and optimize the implementation of new target therapies are needed.
Collapse
Affiliation(s)
- Anabela G. Barros
- Department of Medical Oncology, University Hospital of Coimbra, Coimbra, Portugal
| | - Hélder Mansinho
- Hemato Oncology Department, Garcia de Orta Hospital, Almada, Portugal
| | - Nuno Couto
- Digestive Unit, Champalimaud Clinical Centre, Champalimaud Research Centre, Lisbon, Portugal
| | - Manuel R. Teixeira
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center, Porto, Portugal
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Fernanda S. Tonin
- Pharmaceutical Sciences Postgraduate Program, Federal University of Parana, Curitiba, Brazil
- H&TRC–Health & Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | | | - Filipa Duarte-Ramos
- Department of Pharmacy, Pharmacology and Health Technologies, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
- Research Institute for Medicine (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
- EPIUnit, Instituto de Saúde Pública da Universidade do Porto (ISPUP), Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| |
Collapse
|
334
|
Zeng J, Liu B, Lang X, Wang Z, Fan Y, Gao C, Fu D. Clinical efficacy and safety of irreversible electroporation combined with chemotherapy in stage IV pancreatic cancer treatment. J Cancer Res Ther 2024; 20:1357-1361. [PMID: 39206999 DOI: 10.4103/jcrt.jcrt_73_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: 01/10/2024] [Accepted: 06/22/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE This study evaluates the clinical efficacy and safety of irreversible electroporation (IRE) therapy combined with chemotherapy in patients with stage IV pancreatic cancer. METHODS Between September 2021 and November 2023, we enrolled 38 patients with stage IV pancreatic cancer, with 20 receiving IRE plus chemotherapy and 18 receiving only chemotherapy. We recorded the general information of the patients and regularly followed up postoperative IRE-related adverse reactions. Progression-free survival (PFS) and overall survival (OS) were evaluated during follow-up. RESULTS Median OS was longer in the IRE group than in the chemotherapy group. Median PFS was slightly extended with IRE compared to chemotherapy alone. The mean hospital stay for the IRE group was 5.90 ± 0.75 days. Four serious adverse events occurred after IRE. Postoperative pain scores were significantly lower than preoperative scores. CONCLUSION IRE combined with chemotherapy showed clinical effectiveness in stage IV pancreatic cancer treatment, offering potential pain reduction benefits with fewer adverse effects and shorter hospital stays.
Collapse
Affiliation(s)
- Jia Zeng
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Boyu Liu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xu Lang
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhe Wang
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yong Fan
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Chuntao Gao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Dianxun Fu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
335
|
Murakami T, Kimura Y, Imamura M, Nagayama M, Kato T, Kukita K, Yoshida M, Masaki Y, Nakase H, Takemasa I. Predictors of occult metastases in potentially Resectable pancreatic ductal adenocarcinoma. Surg Open Sci 2024; 20:222-229. [PMID: 39156491 PMCID: PMC11327574 DOI: 10.1016/j.sopen.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 08/20/2024] Open
Abstract
Background Patients with resectable (R) or borderline resectable (BR) pancreatic ductal adenocarcinoma (PDAC) sometimes show unexpected liver, peritoneal, and para-aortic lymph node metastases intraoperatively. Despite radical pancreatectomy, a nonnegligible number of patients relapse within 6 months after surgery. The aim of this study was to identify the preoperative predictors of occult metastases (OM), defined as intraoperative distant metastases or within 6 months after pancreatectomy. Materials and methods This study included patients with R and BR PDAC who underwent curative-intent pancreatectomy or staging laparoscopy between 2006 and 2021. Multivariate logistic regression and Cox hazard analyses were performed to identify the preoperative predictors of OM and to assess the impact of these factors on prognosis after pancreatectomy. Results Of the 279 patients, OM was observed intraoperatively in 47 and postoperatively in 34. In the OM group, there were no differences in prognosis between patients who had intraoperative metastases and recurrence within 6 months (median survival time [MST], 18.1 vs. 12.9 months), and between patients who underwent pancreatectomy and those who did not (MST, 13.9 vs. 18.1 months). Preoperative tumor size ≥22 mm (odds ratio [OR], 2.03; 95 % confidence interval [CI], 1.16-3.53; p = 0.013) and preoperative CA19-9 level ≥ 118.8 U/mL (OR, 2.64; 95 % CI, 1.22-5.73; p = 0.014) were significant predictors of OM. Additionally, positive OM predictors were strong independent prognostic factors for overall survival after pancreatectomy (hazard ratio, 2.47; 95 % CI, 1.54-3.98; p < 0.001). Conclusion Multidisciplinary treatment strategies should be considered for patients with predictors of OM to avoid inappropriate surgical interventions.
Collapse
Affiliation(s)
- Takeshi Murakami
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-0061, Japan
| | - Yasutoshi Kimura
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-0061, Japan
| | - Masafumi Imamura
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-0061, Japan
| | - Minoru Nagayama
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-0061, Japan
| | - Toru Kato
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-0061, Japan
| | - Kazuharu Kukita
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-0061, Japan
| | - Makoto Yoshida
- Department of Medical Oncology, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-0061, Japan
| | - Yoshiharu Masaki
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-0061, Japan
| | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-0061, Japan
| | - Ichiro Takemasa
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, Hokkaido, 060-0061, Japan
| |
Collapse
|
336
|
Mayuko O, Tsunenari T, Einama T, Ichio K, Konno F, Kobayashi K, Yonamine N, Takihata Y, Takao M, Nakazawa A, Kajiwara Y, Ueno H, Kishi Y. Pancreatic cancer with liver metastasis maintaining complete response with gemcitabine monotherapy: A case report. Oncol Lett 2024; 28:370. [PMID: 38933809 PMCID: PMC11200152 DOI: 10.3892/ol.2024.14503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/22/2024] [Indexed: 06/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with a poor prognosis, and it has a recurrence rate of >70%, even in resectable cases. The treatment strategy for recurrent PDAC involves systemic chemotherapy, with gemcitabine (GEM) monotherapy historically serving as the standard of care. The present study describes the case of a patient with PDAC and postoperative liver metastases that maintained clinical complete remission (cCR) for >7 years following GEM monotherapy. A 63-year-old woman with upper abdominal pain was diagnosed with resectable PDAC and underwent pancreaticoduodenectomy. The patient was treated with GEM + S-1 as adjuvant chemotherapy for 6 months. Multiple liver metastases were detected 15 months post-operation and the patient was administered GEM alone. After 12 cycles, computed tomography showed cCR and GEM monotherapy was discontinued after 15 cycles. The patient has had no signs or symptoms of recurrence >7 years after the first recurrence. In addition, the present study analyzed PDAC resection specimens from four patients, including this case, to determine the expression levels of hENT1 protein in the tumor tissues. hENT1 is a transmembrane protein that acts as a nucleoside transporter and is a major mediator of GEM uptake into human cells. In the present case, hENT1 staining exhibited low frequency and weak positivity in the central region, whereas a strong positive reaction was observed in nearly all cell membranes at the invasive front of the cancer. The location, intensity, and frequency of hENT1 staining varied among cases. In conclusion, the efficacy of GEM may be predicted prior to treatment by evaluating hENT1 expression.
Collapse
Affiliation(s)
- Ohara Mayuko
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Takazumi Tsunenari
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Takahiro Einama
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Koki Ichio
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Fukumi Konno
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Kazuki Kobayashi
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Naoto Yonamine
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Yasuhiro Takihata
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Mikiya Takao
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Akiko Nakazawa
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Yoshiki Kajiwara
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| |
Collapse
|
337
|
Cecchini M, Salem RR, Robert M, Czerniak S, Blaha O, Zelterman D, Rajaei M, Townsend JP, Cai G, Chowdhury S, Yugawa D, Tseng R, Mejia Arbelaez C, Jiao J, Shroyer K, Thumar J, Kortmansky J, Zaheer W, Fischbach N, Persico J, Stein S, Khan SA, Cha C, Billingsley KG, Kunstman JW, Johung KL, Wiess C, Muzumdar MD, Spickard E, Aushev VN, Laliotis G, Jurdi A, Liu MC, Escobar-Hoyos L, Lacy J. Perioperative Modified FOLFIRINOX for Resectable Pancreatic Cancer: A Nonrandomized Controlled Trial. JAMA Oncol 2024; 10:1027-1035. [PMID: 38900452 PMCID: PMC11190830 DOI: 10.1001/jamaoncol.2024.1575] [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: 08/18/2023] [Accepted: 01/09/2024] [Indexed: 06/21/2024]
Abstract
Importance Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignant tumor, and durable disease control is rare with the current standard of care, even for patients who undergo surgical resection. Objective To assess whether neoadjuvant modified 5-fluorouracil, leucovorin, oxaliplatin, and irinotecan (mFOLFIRINOX) leads to early control of micrometastasis and improves survival. Design, Setting, and Participants This open-label, single-arm, phase 2 nonrandomized controlled trial for resectable PDAC was conducted at the Yale Smilow Cancer Hospital from April 3, 2014, to August 16, 2021. Pancreatic protocol computed tomography was performed at diagnosis to assess surgical candidacy. Data were analyzed from January to July 2023. Interventions Patients received 6 cycles of neoadjuvant mFOLFIRINOX before surgery and 6 cycles of adjuvant mFOLFIRINOX. Whole blood was collected and processed to stored plasma for analysis of circulating tumor DNA (ctDNA) levels. Tumors were evaluated for treatment response and keratin 17 (K17) expression. Main Outcomes and Measures The primary end point was 12-month progression-free survival (PFS) rate. Additional end points included overall survival (OS), ctDNA level, tumor molecular features, and K17 tumor levels. Survival curves were summarized using Kaplan-Meier estimator. Results Of 46 patients who received mFOLFIRINOX, 31 (67%) were male, and the median (range) age was 65 (46-80) years. A total of 37 (80%) completed 6 preoperative cycles and 33 (72%) underwent surgery. A total of 27 patients (59%) underwent resection per protocol (25 with R0 disease and 2 with R1 disease); metastatic or unresectable disease was identified in 6 patients during exploration. Ten patients underwent surgery off protocol. The 12-month PFS was 67% (90% CI, 56.9-100); the median PFS and OS were 16.6 months (95% CI, 13.3-40.6) and 37.2 months (95% CI, 17.5-not reached), respectively. Baseline ctDNA levels were detected in 16 of 22 patients (73%) and in 3 of 17 (18%) after 6 cycles of mFOLFIRINOX. Those with detectable ctDNA levels 4 weeks postresection had worse PFS (hazard ratio [HR], 34.0; 95% CI, 2.6-4758.6; P = .006) and OS (HR, 11.7; 95% CI, 1.5-129.9; P = .02) compared with those with undetectable levels. Patients with high K17 expression had nonsignificantly worse PFS (HR, 2.7; 95% CI, 0.7-10.9; P = .09) and OS (HR, 3.2; 95% CI, 0.8-13.6; P = .07). Conclusions and Relevance This nonrandomized controlled trial met its primary end point, and perioperative mFOLFIRINOX warrants further evaluation in randomized clinical trials. Postoperative ctDNA positivity was strongly associated with recurrence. K17 and ctDNA are promising biomarkers that require additional validation in future prospective studies. Trial Registration ClinicalTrials.gov Identifier: NCT02047474.
Collapse
Affiliation(s)
- Michael Cecchini
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Ronald R. Salem
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Marie Robert
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Suzanne Czerniak
- Department of Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Ondrej Blaha
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Daniel Zelterman
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Moein Rajaei
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Jeffrey P. Townsend
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Guoping Cai
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Sumedha Chowdhury
- Department of Therapeutic Radiology, Yale School of Public Health, New Haven, Connecticut
| | - Deanne Yugawa
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
- Department of Therapeutic Radiology, Yale School of Public Health, New Haven, Connecticut
| | - Robert Tseng
- Department of Therapeutic Radiology, Yale School of Public Health, New Haven, Connecticut
| | - Carlos Mejia Arbelaez
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
- Department of Therapeutic Radiology, Yale School of Public Health, New Haven, Connecticut
| | - Jingjing Jiao
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Kenneth Shroyer
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - Jaykumar Thumar
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Jeremy Kortmansky
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Wajih Zaheer
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Neal Fischbach
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Justin Persico
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Stacey Stein
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Sajid A. Khan
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Charles Cha
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Kevin G. Billingsley
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - John W. Kunstman
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Kimberly L. Johung
- Department of Therapeutic Radiology, Yale School of Public Health, New Haven, Connecticut
| | - Christina Wiess
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Mandar D. Muzumdar
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | | | | | | | | | | | - Luisa Escobar-Hoyos
- Department of Therapeutic Radiology, Yale School of Public Health, New Haven, Connecticut
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Jill Lacy
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
338
|
Wirth T, Nitschmann S. [Treatment of patients with metastatic pancreatic cancer]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2024; 65:853-854. [PMID: 39023783 DOI: 10.1007/s00108-024-01750-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 07/20/2024]
Affiliation(s)
- Thomas Wirth
- Klinik für Gastroenterologie, Hepatologie, Infektiologie und Endokrinologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625, Hannover, Deutschland.
| | | |
Collapse
|
339
|
Pathak P, Weekes CD, Shroff RT. The Sequencing Conundrum in Localized Pancreatic Adenocarcinoma-Progress or Passive Acceptance? JAMA Oncol 2024; 10:1036-1037. [PMID: 38900451 DOI: 10.1001/jamaoncol.2024.0870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Affiliation(s)
| | - Colin D Weekes
- Division of Hematology-Oncology, Massachusetts General Hospital, Boston
| | - Rachna T Shroff
- Division of Hematology/Oncology, University of Arizona Cancer Center, Tucson
| |
Collapse
|
340
|
Park SJ, Kim JH, Choi SY, Joo I. Important Radiologic and Clinical Factors for Predicting Overall Survival in Pancreatic Adenocarcinoma Patients Who Underwent FOLFIRINOX. Pancreas 2024; 53:e553-e559. [PMID: 38530942 DOI: 10.1097/mpa.0000000000002330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
BACKGROUND To predict poor overall survival (OS) in pancreatic adenocarcinoma (PAC) who underwent FOLFIRINOX (5-fluorouracil/leucovorin/irinotecan/oxaliplatin) using clinical and computed tomography (CT) findings. METHODS A total of 189 patients with PAC who received FOLFIRINOX were retrospectively included. Two reviewers assessed CT findings and resectability based on National Comprehensive Cancer Network guidelines. They determined tumor size changes according to Response Evaluation Criteria in Solid Tumors (RECIST 1.1). Delta measurements were performed. Clinical results, such as whether to perform surgery, were also investigated. A Cox proportional hazard model was used to identify significant predictors for OS. A CT-based nomogram was constructed to predict OS. RESULTS Seventy-four patients (39.2%) underwent surgery. For OS, rim enhancement of PAC on baseline CT (hazard ratio [HR], 1.75; 95% confidence interval [CI], 1.10-2.77; P = 0.018), high delta tumor on baseline CT (HR, 2.46; 95% CI, 1.55-3.91; P < 0.001), progressive disease at follow-up CT (HR, 8.89; 95% CI, 2.94-26.87; P < 0.001), and without surgery (HR, 2.81; 95% CI, 1.49-5.30; P = 0.001) were important features related to poor prognosis. The nomogram showed good predictive ability for the survival. CONCLUSION Both clinical and CT findings were useful for predicting OS after FOLFIRINOX in PAC.
Collapse
Affiliation(s)
- Sae-Jin Park
- From the Department of Radiology, Seoul National University Boramae Hospital, Seoul, Korea
| | | | - Seo-Youn Choi
- Department of Radiology, Soonchunhyang University College of Medicine, Bucheon Hospital, 170 Jomaru-ro, Wonmi-gu, Bucheon
| | | |
Collapse
|
341
|
Mao C, He Y, Xu N, Yan H, Zhang N, Cheng G, Jiang H, Chen M, Chen Y, Wang X, Gu Y, Shen P, Zhang G, Yan J, Yang Z, Ding L, Han Z, Wang Z, Zhang J, Zheng W, Wang J, Qin S. A multicenter, prospective, non-interventional real-world study to assess the effectiveness of mecapegfilgrastim in preventing neutropenia in patients with gastrointestinal cancer. Immun Inflamm Dis 2024; 12:e1348. [PMID: 39105572 PMCID: PMC11301656 DOI: 10.1002/iid3.1348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/12/2024] [Accepted: 07/06/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Mecapegfilgrastim, a long-acting granulocyte-colony stimulating factor has been approved for reducing the incidence of infection, particularly febrile neutropenia (FN), in China. OBJECTIVE We conducted a multicenter prospective observational study to examine the safety and effectiveness of mecapegfilgrastim in preventing neutropenia in gastrointestinal patients receiving the chemotherapy, including S-1/capecitabine-based regimens or the fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI)/fluorouracil, leucovorin, and oxaliplatin (FOLFOX)/fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFIRINOX) regimens. METHOD Five hundred and sixty-one gastrointestinal patients from 40 sites across China, between May 2019 and November 2021, were included. The administration of mecapegfilgrastim was prescribed at the discretion of local physicians. RESULTS The most common adverse drug reactions (ADRs) of any grade for all patients was increased white blood cells (2.9%). Grade 3/4 ADRs were observed for anemia (0.2%), decreased white blood cells (0.2%), and decreased neutrophil count (0.2%). Among the 116 patients who received S-1/capecitabine-based chemotherapy throughout all cycles, ADRs of any grade included anemia (1.7%), myalgia (0.9%), and increased alanine aminotransferase (0.9%). No grade 3/4 ADRs were observed. In 414 cycles of patients who underwent S-1/capecitabine-based regimens, only one (0.2%) cycle experienced grade 4 neutropenia. In the FOLFIRINOX, FOLFOXIRI, and FOLFOX chemotherapy regimens, grade 4 neutropenia occurred in one (2.7%) of 37 cycles, four (4.7%) of 85 cycles, and two (1.2%) of 167 cycles, respectively. CONCLUSION In a real-world setting, mecapegfilgrastim has proven effective in preventing severe neutropenia in gastrointestinal patients following chemotherapy. This includes commonly used moderate or high-risk FN regimens or regimens containing S1/capecitabine, all of which have demonstrated favorable efficacy and safety profiles.
Collapse
Affiliation(s)
- Chenyu Mao
- Department of Medical Oncology, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Ye He
- Department of Medical Oncology, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen UniversityGuangzhouChina
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Haijiao Yan
- Department of OncologyThe First People's Hospital of ChangzhouChangzhouChina
| | - Ningling Zhang
- Department of OncologyAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Gang Cheng
- Department of OncologyBozhou People's HospitalBozhouChina
| | - Hua Jiang
- Department of OncologyThe Second People's Hospital of ChangzhoChangzhouChina
| | - Minbin Chen
- Department of OncologyThe First People's Hospital of KunshanKunshanChina
| | - Yong Chen
- Department of Radiology, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xiaoguang Wang
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Hospital of JiaxingJiaxingChina
| | - Yulan Gu
- Department of OncologyChangshu No 2 People's HospitalChangshuChina
| | - Peng Shen
- Department of Medical Oncology, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Guifang Zhang
- Department of Medical OncologyXinxiang Central HospitalXinxiangChina
| | - Jun Yan
- Department of OncologyThe Central Hospital of JiadingShanghaiChina
| | - Zhe Yang
- Department of RadiologyShandong Provincial HospitalJinanChina
| | - Lifang Ding
- Department of OncologyThe People's Hospital of DanyangDanyangChina
| | - Zhengxiang Han
- Depatment of Integrated Traditional Chinese and Western MedicineThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Zhanggui Wang
- Department of RadiologyThe Second People's Hospital of Anhui ProvinceHefeiChina
| | - Junqi Zhang
- Department of OncologyThe Central Hospital of BazhongBazhongChina
| | - Weie Zheng
- Department of Medical OncologyThe People's Hospital of Rui'anRui'anChina
| | - Jufeng Wang
- Depatment of GastroenterologyHenan Cancer HospitalZhengzhouChina
| | - Shukui Qin
- Chief of HospitalNanjing Tianyinshan HospitalNanjingChina
| |
Collapse
|
342
|
Cashman JR, Cashman EA. Effect of PAWI-2 on pancreatic cancer stem cell tumors. Invest New Drugs 2024; 42:353-360. [PMID: 38789849 DOI: 10.1007/s10637-024-01447-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Worldwide, pancreatic cancer (PC) is a major health problem and almost 0.5 million people were diagnosed with PC in 2020. In the United States, more than 64,000 adults will be diagnosed with PC in 2023. PC is highly resistant to currently available treatments and standard of care chemotherapies cause serious side effects. Most PC patients are resistant to clinical therapies. Combination therapy has showed superior efficacy over single-agent treatment. However, most therapy has failed to show a significant improvement in overall survival due to treatment-related toxicity. Developing efficacious clinically useful PC therapies remains a challenge. Herein, we show the efficacy of an innovative pathway modulator, p53-Activator Wnt Inhibitor-2 (PAWI-2) against tumors arising from human pancreatic cancer stem cells (i.e., hPCSCs, FGβ3 cells). PAWI-2 is a potent inhibitor of tumor growth. In the present study, we showed PAWI-2 potently inhibited growth of tumors from hPCSCs in orthopic xenograft models of both male and female mice. PAWI-2 worked in a non-toxic manner to inhibit tumors. Compared to vehicle-treated animals, PAWI-2 modulated molecular regulators of tumors. Anti-cancer results showed PAWI-2 in vivo efficacy could be correlated to in vitro potency to inhibit FGβ3 cells. PAWI-2 represents a safe, new approach to combat PC.
Collapse
Affiliation(s)
- John R Cashman
- Human BioMolecular Research Institute, San Diego, 5310 Eastgate Mall, San Diego, CA, 92121, USA.
| | - Emily A Cashman
- Human BioMolecular Research Institute, San Diego, 5310 Eastgate Mall, San Diego, CA, 92121, USA
| |
Collapse
|
343
|
Weisz Ejlsmark M, Bahij R, Schytte T, Rønn Hansen C, Bertelsen A, Mahmood F, Bau Mortensen M, Detlefsen S, Weber B, Bernchou U, Pfeiffer P. Adaptive MRI-guided stereotactic body radiation therapy for locally advanced pancreatic cancer - A phase II study. Radiother Oncol 2024; 197:110347. [PMID: 38815694 DOI: 10.1016/j.radonc.2024.110347] [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: 12/01/2023] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
PURPOSE Stereotactic body radiotherapy (SBRT) has emerged as a promising new modality for locally advanced pancreatic cancer (LAPC). The current study evaluated the efficacy and toxicity of SBRT in patients with LAPC (NCT03648632). METHODS This prospective single institution phase II study recruited patients with histologically or cytologically proven adenocarcinoma of the pancreas after more than two months of combination chemotherapy with no sign of progressive disease. Patients were prescribed 50-60 Gy in 5-8 fractions. Patients were initially treated on a standard linac (n = 4). Since 2019, patients were treated using online magnetic resonance (MR) image-guidance on a 1.5 T MRI-linac, where the treatment plan was adapted to the anatomy of the day. The primary endpoint was resection rate. RESULTS Twenty-eight patients were enrolled between August 2018 and March 2022. All patients had non-resectable disease at time of diagnosis. Median follow-up from inclusion was 28.3 months (95 % CI 24.0-NR). Median progression-free and overall survival from inclusion were 7.8 months (95 % CI 5.0-14.8) and 16.5 months (95 % CI 10.7-22.6), respectively. Six patients experienced grade III treatment-related adverse events (jaundice, nausea, vomiting and/or constipation). One of the initial four patients receiving treatment on a standard linac experienced a grade IV perforation of the duodenum. Six patients (21 %) underwent resection. A further one patient was offered resection but declined. CONCLUSION This study demonstrates that SBRT in patients with LAPC was associated with promising overall survival and resection rates. Furthermore, SBRT was safe and well tolerated, with limited severe toxicities.
Collapse
Affiliation(s)
- Mathilde Weisz Ejlsmark
- Department of Oncology, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark.
| | - Rana Bahij
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Tine Schytte
- Department of Oncology, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Christian Rønn Hansen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Laboratory of Radiation Physics, Department of Oncology, Odense University Hospital, Odense, Denmark; Danish Centre of Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Bertelsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Laboratory of Radiation Physics, Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Faisal Mahmood
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Laboratory of Radiation Physics, Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Michael Bau Mortensen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Surgery, Odense University Hospital, Odense, Denmark; Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark
| | - Sönke Detlefsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Pathology, Odense University Hospital, Odense, Denmark; Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark
| | - Britta Weber
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark; Danish Centre of Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Uffe Bernchou
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Laboratory of Radiation Physics, Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark
| |
Collapse
|
344
|
Funo T, Hashimoto D, Yamaki S, Matsumura K, Miyazaki H, Matsui Y, Tsybulskyi D, Sang NT, Yaolin X, Satoi S. Conversion surgery for BRCA-mutated pancreatic ductal adenocarcinoma with liver metastasis treated with platinum-based chemotherapy followed by olaparib. Surg Case Rep 2024; 10:179. [PMID: 39078424 PMCID: PMC11289193 DOI: 10.1186/s40792-024-01975-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/13/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND With recent dramatic developments in chemotherapy, attempts to incorporate surgery into the multidisciplinary treatment of unresectable pancreatic ductal adenocarcinoma with metastasis (UR-M PDAC) have emerged. Maintenance therapy with olaparib after chemotherapy including a platinum-based regimen, which inhibits the poly ADP-ribose polymerase (PARP) involved in DNA repair, was approved for UR-M PDAC with positive BRCA mutations. CASE PRESENTATION A 47-year-old male patient with a high carbohydrate antigen 19-9 (CA19-9) level was diagnosed with PDAC in the pancreatic tail. Staging laparoscopy revealed occult liver metastasis. Because BRCA2 mutation was confirmed, triple combination chemotherapy with SOXIRI (S-1/oxaliplatin/irinotecan) was introduced and continued for 16 weeks, followed by 14 weeks of olaparib. After that, CA19-9 was normalized, and no obvious liver metastases of any size could be seen on imaging studies during chemotherapy. Since staging laparoscopy after chemotherapy proved that the liver metastasis had disappeared, laparoscopic distal pancreatectomy was performed, and curative resection was completed. After adjuvant chemotherapy with olaparib for 12 months, the patient is alive 36 months from his initial diagnosis and 27 months postoperatively without recurrence. CONCLUSION We report a case of PDAC with liver metastasis and BRCA mutation-positivity who underwent conversion surgery and achieved long-term survival after irinotecan-based chemotherapy followed by maintenance therapy with olaparib.
Collapse
Affiliation(s)
- Takumi Funo
- Department of Pancreatobiliary Surgery, Kansai Medical University, 2-5-1 Shin-Machi, Hirakata City, Osaka, 573-1010, Japan
| | - Daisuke Hashimoto
- Department of Pancreatobiliary Surgery, Kansai Medical University, 2-5-1 Shin-Machi, Hirakata City, Osaka, 573-1010, Japan
| | - So Yamaki
- Department of Pancreatobiliary Surgery, Kansai Medical University, 2-5-1 Shin-Machi, Hirakata City, Osaka, 573-1010, Japan
| | - Kazuki Matsumura
- Department of Pancreatobiliary Surgery, Kansai Medical University, 2-5-1 Shin-Machi, Hirakata City, Osaka, 573-1010, Japan
| | - Hidetaka Miyazaki
- Department of Pancreatobiliary Surgery, Kansai Medical University, 2-5-1 Shin-Machi, Hirakata City, Osaka, 573-1010, Japan
| | - Yuki Matsui
- Department of Pancreatobiliary Surgery, Kansai Medical University, 2-5-1 Shin-Machi, Hirakata City, Osaka, 573-1010, Japan
| | - Denys Tsybulskyi
- Department of Pancreatobiliary Surgery, Kansai Medical University, 2-5-1 Shin-Machi, Hirakata City, Osaka, 573-1010, Japan
| | - Nguyen Thanh Sang
- Department of Pancreatobiliary Surgery, Kansai Medical University, 2-5-1 Shin-Machi, Hirakata City, Osaka, 573-1010, Japan
| | - Xu Yaolin
- Department of Pancreatobiliary Surgery, Kansai Medical University, 2-5-1 Shin-Machi, Hirakata City, Osaka, 573-1010, Japan
| | - Sohei Satoi
- Department of Pancreatobiliary Surgery, Kansai Medical University, 2-5-1 Shin-Machi, Hirakata City, Osaka, 573-1010, Japan.
- Division of Surgical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
345
|
Rincon-Torroella J, Dal Molin M, Mog B, Han G, Watson E, Wyhs N, Ishiyama S, Ahmedna T, Minn I, Azad NS, Bettegowda C, Papadopoulos N, Kinzler KW, Zhou S, Vogelstein B, Gabrielson K, Sur S. ME3BP-7 is a targeted cytotoxic agent that rapidly kills pancreatic cancer cells expressing high levels of monocarboxylate transporter MCT1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.23.550207. [PMID: 37546808 PMCID: PMC10401962 DOI: 10.1101/2023.07.23.550207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Nearly 30% of Pancreatic ductal adenocarcinoma (PDAC)s exhibit a marked overexpression of Monocarboxylate Transporter 1 (MCT1) offering a unique opportunity for therapy. However, biochemical inhibitors of MCT1 have proven unsuccessful in clinical trials. In this study we present an alternative approach using 3-Bromopyruvate (3BP) to target MCT1 overexpressing PDACs. 3BP is a cytotoxic agent that is known to be transported into cells via MCT1, but its clinical usefulness has been hampered by difficulties in delivering the drug systemically. We describe here a novel microencapsulated formulation of 3BP (ME3BP-7), that is effective against a variety of PDAC cells in vitro and remains stable in serum. Furthermore, systemically administered ME3BP-7 significantly reduces pancreatic cancer growth and metastatic spread in multiple orthotopic models of pancreatic cancer with manageable toxicity. ME3BP-7 is, therefore, a prototype of a promising new drug, in which the targeting moiety and the cytotoxic moiety are both contained within the same single small molecule. One Sentence Summary ME3BP-7 is a novel formulation of 3BP that resists serum degradation and rapidly kills pancreatic cancer cells expressing high levels of MCT1 with tolerable toxicity in mice.
Collapse
|
346
|
Imamura T, Komatsu S, Nishibeppu K, Kiuchi J, Ohashi T, Konishi H, Shiozaki A, Yamamoto Y, Moriumura R, Ikoma H, Ochiai T, Otsuji E. Urinary microRNA-210-3p as a novel and non-invasive biomarker for the detection of pancreatic cancer, including intraductal papillary mucinous carcinoma. BMC Cancer 2024; 24:907. [PMID: 39069624 DOI: 10.1186/s12885-024-12676-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: 08/18/2023] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND This study aims to explore novel microRNAs in urine for screening and predicting clinical characteristics in pancreatic cancer (PC) patients using a microRNA array-based approach. METHODS We used the Toray® 3D-Gene microRNA array-based approach to compare urinary levels between PC patients and healthy volunteers. RESULTS (1) Four oncogenic microRNAs (miR-744-5p, miR-572, miR-210-3p, and miR-575) that were highly upregulated in the urine of PC patients compared to healthy individuals were identified by comprehensive microRNA array analysis. (2) Test-scale analysis by quantitative RT-PCR for each group of 20 cases showed that miR-210-3p was significantly upregulated in the urine of PC patients compared to healthy individuals (P = 0.009). (3) Validation analysis (58 PC patients and 35 healthy individuals) confirmed that miR-210-3p was significantly upregulated in the urine of PC patients compared to healthy individuals (P < 0.001, area under the receiver operating characteristic curve = 0.79, sensitivity: 0.828, specificity: 0.743). We differentiated PC patients into invasive ductal carcinoma (IDCa) and intraductal papillary mucinous carcinoma (IPMC) groups. In addition to urinary miR-210-3p levels being upregulated in IDCa over healthy individuals (P = 0.009), urinary miR-210-3p levels were also elevated in IPMC over healthy individuals (P = 0.0018). Urinary miR-210-3p can differentiate IPMC from healthy individuals by a cutoff of 8.02 with an AUC value of 0.762, sensitivity of 94%, and specificity of 63%. (4) To test whether urinary miR210-3p levels reflected plasma miR-210-3p levels, we examined the correlation between urinary and plasma levels. Spearman's correlation analysis showed a moderate positive correlation (ρ = 0.64, P = 0.005) between miR-210-3p expression in plasma and urine. CONCLUSIONS Urinary miR-210-3p is a promising, non-invasive diagnostic biomarker of PC, including IPMC. TRIAL REGISTRATION Not applicable.
Collapse
MESH Headings
- Humans
- MicroRNAs/urine
- MicroRNAs/blood
- MicroRNAs/genetics
- Female
- Male
- Biomarkers, Tumor/urine
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/blood
- Pancreatic Neoplasms/urine
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/blood
- Middle Aged
- Aged
- Adenocarcinoma, Mucinous/urine
- Adenocarcinoma, Mucinous/genetics
- Adenocarcinoma, Mucinous/diagnosis
- ROC Curve
- Case-Control Studies
- Gene Expression Regulation, Neoplastic
- Adult
- Carcinoma, Pancreatic Ductal/urine
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/blood
Collapse
Affiliation(s)
- Taisuke Imamura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Keiji Nishibeppu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Jun Kiuchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yusuke Yamamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Ryo Moriumura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hisashi Ikoma
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Toshiya Ochiai
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
347
|
de Jesus VHF, Donadio MDS, de Brito ÂBC, Gentilli AC. A narrative review on rare types of pancreatic cancer: should they be treated as pancreatic ductal adenocarcinomas? Ther Adv Med Oncol 2024; 16:17588359241265213. [PMID: 39072242 PMCID: PMC11282540 DOI: 10.1177/17588359241265213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 06/13/2024] [Indexed: 07/30/2024] Open
Abstract
Pancreatic cancer is one of the deadliest malignancies in humans and it is expected to play a bigger part in cancer burden in the years to come. Pancreatic ductal adenocarcinoma (PDAC) represents 85% of all primary pancreatic malignancies. Recently, much attention has been given to PDAC, with significant advances in the understanding of the mechanisms underpinning disease initiation and progression, along with noticeable improvements in overall survival in both localized and metastatic settings. However, given their rarity, rare histological subtypes of pancreatic cancer have been underappreciated and are frequently treated as PDAC, even though they might present non-overlapping molecular alterations and clinical behavior. While some of these rare histological subtypes are true variants of PDAC that should be treated likewise, others represent separate clinicopathological entities, warranting a different therapeutic approach. In this review, we highlight clinical, pathological, and molecular aspects of rare histological types of pancreatic cancer, along with the currently available data to guide treatment decisions.
Collapse
Affiliation(s)
- Victor Hugo Fonseca de Jesus
- Oncoclínicas, Department of Gastrointestinal Medical Oncology, Santos Dumont St. 182, 4 floor, Florianópolis, Santa Catarina 88015-020, Brazil
- Department of Medical Oncology, Centro de Pesquisas Oncológicas, Florianópolis, Santa Catarina, Brazil
| | | | | | | |
Collapse
|
348
|
Zhou S, Ze X, Feng D, Liu L, Shi Y, Yu M, Huang L, Wang Y, Men H, Wu J, Yuan Z, Zhou M, Xu J, Li X, Yao H. Identification of 6-Fluorine-Substituted Coumarin Analogues as POLRMT Inhibitors with High Potency and Safety for Treatment of Pancreatic Cancer. J Med Chem 2024. [PMID: 39049433 DOI: 10.1021/acs.jmedchem.4c01178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Increasing evidence has demonstrated that oxidative phosphorylation (OXPHOS) is closely associated with the progression of pancreatic cancer (PC). Given its central role in mitochondrial transcription, the human mitochondrial RNA polymerase (POLRMT) is a promising target for developing PC treatments. Herein, structure-activity relationship exploration led to the identification of compound S7, which was the first reported POLRMT inhibitor possessing single-digit nanomolar potency of inhibiting PC cells proliferation. Mechanistic studies showed that compound S7 exerted antiproliferative effects without affecting the cell cycle, apoptosis, mitochondrial membrane potential (MMP), or intracellular reactive oxygen species (ROS) levels specifically in MIA PaCa-2 cells. Notably, compound S7 inhibited tumor growth in MIA PaCa-2 xenograft tumor model with a tumor growth inhibition (TGI) rate of 64.52% demonstrating significant improvement compared to the positive control (44.80%). In conclusion, this work enriched SARs of POLRMT inhibitors, and compound S7 deserved further investigations of drug-likeness as a candidate for PC treatment.
Collapse
Affiliation(s)
- Shengnan Zhou
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Xiaotong Ze
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Dazhi Feng
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Lihua Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Yuning Shi
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Minghui Yu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Lijuan Huang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Yunyue Wang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Hanlu Men
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Jianbing Wu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Zhenwei Yuan
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Mengze Zhou
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Longmian Avenue 639, Nanjing 211198, China
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Xinnan Li
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Hong Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| |
Collapse
|
349
|
Shibuki T, Otsuka T, Shimokawa M, Nakazawa J, Arima S, Fukahori M, Miwa K, Okabe Y, Koga F, Ueda Y, Kubotsu Y, Makiyama A, Shimokawa H, Takeshita S, Nishikawa K, Komori A, Otsu S, Hosokawa A, Sakai T, Oda H, Kawahira M, Arita S, Honda T, Taguchi H, Tsuneyoshi K, Kawaguchi Y, Fujita T, Sakae T, Nio K, Ide Y, Ureshino N, Shirakawa T, Mizuta T, Mitsugi K. Nanoliposomal irinotecan with fluorouracil and folinic acid, FOLFIRINOX, and S-1 as second-line treatment for unresectable pancreatic cancer after gemcitabine/nab-paclitaxel. Sci Rep 2024; 14:16906. [PMID: 39043707 PMCID: PMC11266600 DOI: 10.1038/s41598-024-65689-8] [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: 11/14/2023] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
This study aimed to compare second-line treatment outcomes for patients with unresectable pancreatic cancer previously treated with gemcitabine plus nab-paclitaxel (GnP) therapy. We conducted an integrated analysis of two retrospective studies included 318 patients receiving nanoliposomal irinotecan + 5-fluorouracil/folinic acid (NFF) (n = 102), S-1 (n = 57), or FOLFIRINOX (n = 14) as second-line treatment. Median overall survival (OS) in the NFF group was 9.08 months, significantly better than S-1 (4.90 months, P = 0.002). FOLFIRINOX had a median OS of 4.77 months, not statistically different from NFF. Subgroup analyses of OS indicated NFF was generally superior, however, a statistical interaction was observed between the treatment regimen in serum Alb < 3.5 g/dL (P = 0.042) and serum CRP ≥ 0.3 mg/dL (P = 0.006). Median progression-free survival (PFS) was 2.93 months for NFF, significantly better than S-1 (2.53 months, P = 0.024), while FOLFIRINOX had a comparable PFS (3.04 months, P = 0.948). Multivariate analysis identified the serum CRP, serum CA19-9, duration of first-line GnP therapy, and use (yes/no) of S-1 for second-line treatment as independent predictors for OS. This study concludes that second-line NFF therapy demonstrated a more favorable OS compared to S-1 therapy, however, it is still important to consider the patient background characteristics while selecting the most appropriate treatment.
Collapse
Affiliation(s)
- Taro Shibuki
- Division of Drug and Diagnostic Development Promotion, Department for the Promotion of Drug and Diagnostic Development, Translational Research Support Office, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
| | - Taiga Otsuka
- Department of Medical Oncology, Saga-Ken Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga, 840-8571, Japan.
- Department of Internal Medicine, Minato Medical Clinic, 3-11-3 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka, 810-0072, Japan.
| | - Mototsugu Shimokawa
- Clinical Research Institute, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka-shi, Fukuoka, 811-1395, Japan
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube-shi, Yamaguchi, 755-8505, Japan
| | - Junichi Nakazawa
- Department of Gastroenterology, Kagoshima City Hospital, 37-1 Uearata-cho, Kagoshima-shi, Kagoshima, 890-8760, Japan
| | - Shiho Arima
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima-shi, Kagoshima, 890-8520, Japan
| | - Masaru Fukahori
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume-shi, Fukuoka, 830-0011, Japan.
- Kyoto Innovation Center for Next Generation Clinical Trials and iPS Cell Therapy, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Keisuke Miwa
- Multidisciplinary Treatment Cancer Center, Kurume University Hospital, 67 Asahi-machi, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Yoshinobu Okabe
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Futa Koga
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga, 840-8571, Japan
| | - Yujiro Ueda
- Department of Hematology and Oncology, Japanese Red Cross Kumamoto Hospital, 2-1-1 Nagamine-Minami, Higashi-ku, Kumamoto-shi, Kumamoto, 861-8520, Japan
| | - Yoshihito Kubotsu
- Department of Internal Medicine, Karatsu Red Cross Hospital, 2430 Watada,Karatsu-shi, Saga, 847-8588, Japan
| | - Akitaka Makiyama
- Department of Hematology Oncology, Japan Community Healthcare Organization Kyushu Hospital, 1-8-1 Kishinoura, Yahatanishi-ku, Kitakyushu-shi, Fukuoka, 806-8501, Japan
- Cancer Center, Gifu University Hospital, 1-1 Yanagido, Gifu-shi, Gifu, 501-1194, Japan
| | - Hozumi Shimokawa
- Department of Hematology Oncology, Japan Community Healthcare Organization Kyushu Hospital, 1-8-1 Kishinoura, Yahatanishi-ku, Kitakyushu-shi, Fukuoka, 806-8501, Japan
| | - Shigeyuki Takeshita
- Department of Gastroenterology, Japanese Red Cross Nagasaki Genbaku Hospital, 3-15, Morimachi, Nagasaki-shi, Nagasaki, 852-8511, Japan
| | - Kazuo Nishikawa
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-shi, Oita, 879-5593, Japan
| | - Azusa Komori
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-shi, Oita, 879-5593, Japan
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, 160 Minamiumemoto-cho, Matsuyama-shi, Ehime, 791-0280, Japan
| | - Satoshi Otsu
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-shi, Oita, 879-5593, Japan
| | - Ayumu Hosokawa
- Department of Clinical Oncology, University of Miyazaki Hospital, 5200, Kiyotakechoukihara, Miyazaki-shi, Miyazaki, 889-1692, Japan
| | - Tatsunori Sakai
- Department of Medical Oncology, National Hospital Organization Kumamoto Medical Center, 1-5, Ninomaru, Chuo-ku, Kumamoto-shi, Kumamoto, 860-0008, Japan
| | - Hisanobu Oda
- Division of Integrative Medical Oncology, Saiseikai Kumamoto Hospital, 5-3-1,Oumi, Minami-ku, Kmamoto-shi, Kumamoto, 861-4193, Japan
| | - Machiko Kawahira
- Department of Gastroenterology, Kagoshima Kouseiren Hospital, 1-13-1, Yojirou,Kagoshima-shi, Kagoshima, 890-0062, Japan
| | - Shuji Arita
- Department of Chemotherapy, Miyazaki Prefectural Miyazaki Hospital, Miyazaki, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8501, Japan
| | - Hiroki Taguchi
- Department of Gastroenterology, Kagoshima City Hospital, 37-1 Uearata-cho, Kagoshima-shi, Kagoshima, 890-8760, Japan
- Department of Gastroenterology, Saiseikai Sendai Hospital, 2-46 Harada-cho, Satsumasendai-shi, Kagoshima, 895-0074, Japan
| | - Kengo Tsuneyoshi
- Department of Gastroenterology, Izumi General Medical Center, 520, Myoujin-cho, Izumi-shi, Kagoshima, 899-0131, Japan
| | - Yasunori Kawaguchi
- Department of Gastroenterology, Asakura Medical Association Hospital, 422-1, Raiha, Asakura-shi, Fukuoka, 838-0069, Japan
| | - Toshihiro Fujita
- Department of Gastroenterology, Saiseikai Sendai Hospital, 2-46 Harada-cho, Satsumasendai-shi, Kagoshima, 895-0074, Japan
| | - Takahiro Sakae
- Department of Gastroenterology, Saiseikai Sendai Hospital, 2-46 Harada-cho, Satsumasendai-shi, Kagoshima, 895-0074, Japan
| | - Kenta Nio
- Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-cho, Sasebo-shi, Nagasaki, 857-8575, Japan
- Department of Medical Oncology, Hamanomachi Hospital, 3-3-1 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka, 810-8539, Japan
| | - Yasushi Ide
- Department of Internal Medicine, Karatsu Red Cross Hospital, 2430 Watada,Karatsu-shi, Saga, 847-8588, Japan
- Department of Internal Medicine, National Hospital Organization Saga Hospital, 1-20-1 Hinode, Saga-shi, Saga, 849-8577, Japan
| | - Norio Ureshino
- Department of Medical Oncology, Saga-Ken Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga, 840-8571, Japan
- Department of Medical Oncology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba, 292-8535, Japan
| | - Tsuyoshi Shirakawa
- Eikoh Hospital, 3-8-15 Befu-nishi, Shime-machi, Kasuya-gun, Fukuoka, 811-2232, Japan
- Clinical Hematology Oncology Treatment Study Group, 1-14-6 Muromi-gaoka, Nishi-ku, Fukuoka-shi, Fukuoka, 819-0030, Japan
| | - Toshihiko Mizuta
- Department of Internal Medicine, Fujikawa Hospital, 1-2-6 Matsubara, Saga-shi, Saga, 840-0831, Japan
| | - Kenji Mitsugi
- Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-cho, Sasebo-shi, Nagasaki, 857-8575, Japan
- Department of Medical Oncology, Hamanomachi Hospital, 3-3-1 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka, 810-8539, Japan
| |
Collapse
|
350
|
Burke CE, Eng NL, Yee NS, Peng JS. Complete response to chemotherapy in a 6-year survivor of metastatic pancreatic cancer. BMJ Case Rep 2024; 17:e261008. [PMID: 39038872 DOI: 10.1136/bcr-2024-261008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024] Open
Abstract
A woman in her 40s underwent evaluation for abdominal pain, jaundice and acholic stools and was diagnosed with metastatic pancreatic head adenocarcinoma. She was enrolled in a clinical trial investigating the benefits of ibrutinib with nab-paclitaxel and gemcitabine, and subsequently received modified FOLFIRINOX. Over the course of 6 years on chemotherapy, she experienced complete regression of the pancreatic and liver lesions, as well as normalisation of her tumour markers. She has been off chemotherapy for 6 months with no evidence of disease and normal tumour markers. Despite advances in chemotherapy and surgical options, metastatic pancreatic adenocarcinoma continues to carry a grim prognosis. This case report demonstrates a rare case of a long-term survivor of unresectable metastatic pancreatic adenocarcinoma treated with chemotherapy alone.
Collapse
Affiliation(s)
| | - Nina L Eng
- Department of Surgery, Penn State College of Medicine, Hershey, PA, USA
| | - Nelson S Yee
- Division of Hematology and Oncology, Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - June S Peng
- Divsion of Surgical Oncology, Department of Surgery, Penn State College of Medicine, Hershey, PA, USA
- Division of Surgical Oncology, Department of Surgery, UCSF, San Francisco, CA, USA
| |
Collapse
|