1
|
Calheiros J, Silva R, Barbosa F, Morais J, Moura SR, Almeida S, Fiorini E, Mulhovo S, Aguiar TQ, Wang T, Ricardo S, Almeida MI, Domingues L, Melo SA, Corbo V, Ferreira MJU, Saraiva L. A first-in-class inhibitor of homologous recombination DNA repair counteracts tumour growth, metastasis and therapeutic resistance in pancreatic cancer. J Exp Clin Cancer Res 2025; 44:129. [PMID: 40275348 PMCID: PMC12020112 DOI: 10.1186/s13046-025-03389-5] [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: 12/27/2024] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is among the cancer types with poorest prognosis and survival rates primarily due to resistance to standard-of-care therapies, including gemcitabine (GEM) and olaparib. Particularly, wild-type (wt)BRCA tumours, the most prevalent in PDAC, are more resistant to DNA-targeting agents like olaparib, restraining their clinical application. Recently, we disclosed a monoterpene indole alkaloid derivative (BBIT20) as a new inhibitor of homologous recombination (HR) DNA repair with anticancer activity in breast and ovarian cancer. Since inhibition of DNA repair enhances the sensitivity of cancer cells to chemotherapy, we aimed to investigate the anticancer potential of BBIT20 against PDAC, particularly carrying wtBRCA. METHODS In vitro and in vivo PDAC models, particularly human cell lines (including GEM-resistant PDAC cells), patient-derived organoids and xenograft mice of PDAC were used to evaluate the anticancer potential of BBIT20, alone and in combination with GEM or olaparib. Disruption of the BRCA1-BARD1 interaction by BBIT20 was assessed by co-immunoprecipitation, immunofluorescence and yeast two-hybrid assay. RESULTS The potent antiproliferative activity of BBIT20, superior to olaparib, was demonstrated in PDAC cells regardless of BRCA status, by inducing cell cycle arrest, apoptosis, and DNA damage, while downregulating HR. The disruption of DNA double-strand breaks repair by BBIT20 was further reinforced by non-homologous end joining (NHEJ) suppression. The inhibition of BRCA1-BARD1 heterodimer by BBIT20 was demonstrated in PDAC cells and confirmed in a yeast two-hybrid assay. In GEM-resistant PDAC cells, BBIT20 showed potent antiproliferative, anti-migratory and anti-invasive activity, overcoming GEM resistance by inhibiting the multidrug resistance P-glycoprotein, upregulating the intracellular GEM-transporter ENT1, and downregulating GEM resistance-related microRNA-20a and GEM metabolism enzymes as RRM1/2. Furthermore, BBIT20 did not induce resistance in PDAC cells. It inhibited the growth of patient-derived PDAC organoids, by inducing apoptosis, repressing HR, and potentiating olaparib and GEM cytotoxicity. The enhancement of olaparib antitumor activity by BBIT20 was confirmed in xenograft mice of PDAC. Notably, it hindered tumour growth and liver metastasis formation, improving survival of orthotopic xenograft mice of PDAC. Furthermore, its potential as a stroma-targeting agent, reducing fibrotic extracellular matrix and overcoming desmoplasia, associated with an enhancement of immune cell response by depleting PD-L1 expression in tumour tissues, renders BBIT20 even more appealing for combination therapy, particularly with immunotherapy. CONCLUSION These findings underscore the great potential of BBIT20 as a novel multifaceted anticancer drug candidate for PDAC treatment.
Collapse
Grants
- 2020.04613.BD FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2020.06020.BD FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- AIRC; IG No 288801 Associazione Italiana Ricerca sul Cancro
- AIRC; IG No 288801 Associazione Italiana Ricerca sul Cancro
- NHI; HHSN26100008 NCI NIH HHS
- National Cancer Institute
Collapse
Affiliation(s)
- Juliana Calheiros
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Rita Silva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Filipa Barbosa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, 1649-003, Portugal
| | - João Morais
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Sara Reis Moura
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Sofia Almeida
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Elena Fiorini
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Silva Mulhovo
- Centro de Estudos Moçambicanos e de Etnociências (CEMEC), Faculty of Natural Sciences and Mathematics, Pedagogical University, Maputo, 21402161, Mozambique
| | - Tatiana Q Aguiar
- CEB - Centre of Biological Engineering, University of Minho, Braga, 4710-057, Portugal
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Tao Wang
- Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, China
| | - Sara Ricardo
- Associate Laboratory i4HB - Institute for Health and Bioeconomy and UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, 4585-116, Portugal
| | - Maria Inês Almeida
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Lucília Domingues
- CEB - Centre of Biological Engineering, University of Minho, Braga, 4710-057, Portugal
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Sonia A Melo
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
- Department of Pathology, Faculty of Medicine University of Porto, Al. Prof. Hernâni Monteiro, Porto, 4200-319, Portugal
- Porto Comprehensive Cancer Centre (P.CCC) Raquel Seruca, Porto, Portugal
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Maria-José U Ferreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, 1649-003, Portugal.
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal.
| |
Collapse
|
2
|
Farooq AR, Zhang AX, Chan-Seng-Yue M, Topham JT, O'Kane GM, Jang GH, Fischer S, Dodd A, Holter S, Wilson J, Grant RC, Aung KL, Zogopoulos G, Elimova E, Prince R, Jang R, Moore M, Biagi J, Tang P, Goodwin R, Bathe OF, Marra M, Laskin J, Renouf DJ, Schaeffer DF, Karasinska JM, Notta F, Gallinger S, Knox JJ, Tsang ES. The tandem duplicator phenotype may be a novel targetable subgroup in pancreatic cancer. NPJ Precis Oncol 2025; 9:100. [PMID: 40185871 PMCID: PMC11971333 DOI: 10.1038/s41698-025-00888-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Tandem duplicator phenotype (TDP) consists of distinct genomic rearrangements where tandem duplications are randomly distributed. In this study, we characterized the prevalence and outcomes of TDP in a large series of prospectively sequenced tumors from patients with pancreatic ductal adenocarcinomas (PDAC). Whole-genome sequencing (WGS) was performed in 530 PDAC cases from the PanCuRx Initiative, COMPASS and PanGen/POG trials in Canada. Of 530 cases, 52 were identified as TDP (9.8%; 13 resected, 39 advanced). Etiological subgroups of TDP included BRCA1 (n = 9), CCNE1 (n = 4), and unknown (n = 39). Presence of TDP was not prognostic in resected specimens (p = 0.77) compared with non-HRD and non-TDP cases, described as typicals. In advanced cases, when stratified for only classical subtype cases, platinum therapy was correlated with longer response in non-BRCA1 TDP vs. typicals (p = 0.0036). There was no difference in overall survival between TDP and typicals (p = 0.5).TDP represents a potential novel targetable subgroup for chemotherapy selection in PDAC.
Collapse
Affiliation(s)
| | - Amy X Zhang
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | - James T Topham
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | | | - Gun Ho Jang
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | - Anna Dodd
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Spring Holter
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Julie Wilson
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | | | | | - Elena Elimova
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | | - Raymond Jang
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Malcolm Moore
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - James Biagi
- Cancer Centre of Southeastern Ontario/Queen's University, Kingston, ON, Canada
| | - Patricia Tang
- Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Rachel Goodwin
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital, The University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Oliver F Bathe
- Department of Oncology, Tom Baker Cancer Center, University of Calgary, 1331 29th St NW, Calgary, AB, T2N 4N2, Canada
| | - Marco Marra
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - Janessa Laskin
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - Daniel J Renouf
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - David F Schaeffer
- Department of Pathology, Vancouver General Hospital, Vancouver, BC, Canada
| | | | - Faiyaz Notta
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | | | - Erica S Tsang
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
3
|
Gilani S, Ibrahim MA, Mujeeb Q, Khir I. Pancreatic carcinosarcoma: a rare type of pancreatic neoplasia with long-term survival. BMJ Case Rep 2025; 18:e262648. [PMID: 40107743 DOI: 10.1136/bcr-2024-262648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
Abstract
A woman in her 60s presented with persistent abdominal pain and jaundice. She had a history of BRCA1 gene mutation and was previously treated for triple-negative early breast cancer. Abdominal ultrasound and CT scan of the abdomen revealed a mass around the pancreatic head. Magnetic resonance cholangiopancreatography showed a dilated common bile duct and a normal pancreatic duct. Liver function improved after endoscopic retrograde cholangiopancreatography and biliary stent placement. Staging CT confirmed no distant metastasis. The patient underwent Whipple's procedure to remove the pancreatic lesion. Histology from the resection confirmed carcinosarcoma with positive margins, along with lymphovascular, perineural and nodal involvement. She was offered adjuvant chemotherapy with a combination of Folinic acid, 5-fluorouracil, Oxaliplatin and Irinotecan (FOLFIRINOX). Due to derangement in liver enzymes after one cycle of chemotherapy, an MRI scan confirmed liver metastasis. The same chemotherapy regimen was continued in a palliative setting, initially showing a partial response to the liver metastases, but subsequently resulting in a long-term complete radiological response. Pancreatic carcinosarcoma is a rare type of cancer with no clear consensus on the appropriate management. In this case, FOLFIRINOX palliative chemotherapy led to long-term disease control. Future studies on molecular profiling may provide insights for genotype-driven, personalised treatment strategies for similar cases.
Collapse
Affiliation(s)
- Shahid Gilani
- Clinical Oncology, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | | | - Qudsia Mujeeb
- University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | - Ibrahim Khir
- University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| |
Collapse
|
4
|
Shao C, Zhou H, Chen C, Dettman EJ, Ren Y, Cristescu R, Gozman A, Jin F. Prevalence of Homologous Recombination Repair Mutations and Association with Clinical Outcomes in Patients with Solid Tumors: A Study Using the AACR Project GENIE Dataset. Cancers (Basel) 2025; 17:577. [PMID: 40002171 PMCID: PMC11852487 DOI: 10.3390/cancers17040577] [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: 12/17/2024] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Mutations in BRCA1 and/or BRCA2 (BRCAm) and other homologous recombination repair genes (HRRm) are associated with several cancers. We evaluated the prevalence and association with overall survival (OS) of somatic BRCAm and HRRm among patients with advanced solid tumors. METHODS We used deidentified data from the AACR GENIE Biopharma Collaborative dataset derived from patients with tumors genotyped using next-generation sequencing between 1 January 2014 and 31 December 2017. Eligible patients were aged ≥18 years diagnosed with non-small-cell lung, colorectal, breast, bladder, prostate, or pancreatic cancer, with documented BRCA/HRR somatic mutation status. The primary analysis was OS (censored at the start of poly[ADP ribose] polymerase inhibitors [PARPi]/immunotherapy) after initiation of second-line therapy since most patients had sequencing after first-line therapy. RESULTS Among eligible patients, 242/7022 (3.4%) had BRCAm and 477/5474 (8.7%) had HRRm. Adjusted OS HRs (95% CI) for the primary analysis (using the initiation of second-line therapy as index date) were 0.79 (0.61-1.03) with/without BRCAm (n = 116/n = 3394) and 0.83 (0.69-0.99) with/without HRRm (n = 247/n = 2656); in sensitivity analysis of patients with stage IV disease, HRs were 0.97 (0.68-1.38) with/without BRCAm (n = 58/n = 1847) and 0.92 (0.73-1.18) with/without HRRm (n = 132/n = 1488). CONCLUSIONS Overall, BRCAm and HRRm did not show a strong association with OS, with a trend toward longer OS among patients receiving standard second-line therapies excluding PARPi/immunotherapy.
Collapse
Affiliation(s)
- Changxia Shao
- Merck & Co., Inc., Rahway, NJ 07065, USA; (H.Z.); (C.C.); (E.J.D.); (Y.R.); (R.C.); (A.G.); (F.J.)
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Xiao M, Tang R, Pan H, Yang J, Tong X, Xu H, Guo Y, Lei Y, Wu D, Lei Y, Han Y, Ma Z, Wang W, Xu J, Yu X, Shi S. TPX2 serves as a novel target for expanding the utility of PARPi in pancreatic cancer through conferring synthetic lethality. Gut 2025; 74:410-423. [PMID: 39500552 PMCID: PMC11874363 DOI: 10.1136/gutjnl-2024-332782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/14/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND PARP inhibitors (PARPi) have been licensed for the maintenance therapy of patients with metastatic pancreatic cancer carrying pathogenic germline BRCA1/2 mutations. However, mutations in BRCA1/2 are notably rare in pancreatic cancer. OBJECTIVE There is a significant unmet clinical need to broaden the utility of PARPi. DESIGN RNA sequencing was performed to screen potential targets for PARPi sensitivity. The synthetic lethal effects were verified in patient-derived xenograft (PDX), xenograft and patient-derived organoid models. Mechanisms were explored via LC‒MS/MS, coimmunoprecipitation, laser microirradiation, immunofluorescence, the homologous recombination (HR) or non-homologous end joining (NHEJ) reporter system, in situ proximity ligation assay and live-cell time-lapse imaging analyses. RESULTS Targeting protein for Xenopus kinesin-like protein 2 (TPX2) is an exploitable vulnerability. TPX2 was downregulated in PDX models sensitive to PARPi, and TPX2 inhibition conferred synthetic lethality to PARPi both in vitro and in vivo. Mechanistically, TPX2 functions in a cell cycle-dependent manner. In the S/G2 phase, ATM-mediated TPX2 S634 phosphorylation promotes BRCA1 recruitment to double-strand breaks (DSBs) for HR repair, whereas non-phosphorylated TPX2 interacts with 53BP1 to recruit it for NHEJ. The balance between phosphorylated and non-phosphorylated TPX2 determines the DSB repair pathway choice. During mitosis, TPX2 phosphorylation enhances Aurora A activity, promoting mitotic progression and chromosomal stability. Targeting TPX2 S634 phosphorylation with a cell-penetrating peptide causes genomic instability and mitotic catastrophe and enhances PARPi sensitivity. Additionally, the inhibition of TPX2 or S634 phosphorylation combined with gemcitabine further sensitised pancreatic cancer to PARPi. CONCLUSIONS Our findings revealed the dual-functional significance of TPX2 in controlling DNA DSB repair pathway choice and mitotic progression, suggesting a potential therapeutic strategy involving PARPi for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Mingming Xiao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Haoqi Pan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Jing Yang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Xuhui Tong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - He Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Yanmei Guo
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Yalan Lei
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Di Wu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Yubin Lei
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yamei Han
- Department of Biochemistry and Molecular Biology,Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhilong Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Milella M, Orsi G, di Marco M, Salvatore L, Procaccio L, Noventa S, Bozzarelli S, Garajova I, Vasile E, Giordano G, Macchini M, Cavaliere A, Gaule M, Bergamo F, Chiaravalli M, Palloni A, Carloni R, Bittoni A, Niger M, Rapposelli IG, Rodriquenz MG, Scartozzi M, Mosconi S, Giommoni E, Bernardini I, Paratore C, Spallanzani A, Bencardino K, Forti L, Tamburini E, Lonardi S, Scarpa A, Cascinu S, Tortora G, Sperduti I, Reni M. Real-World Impact of Olaparib Exposure in Advanced Pancreatic Cancer Patients Harboring Germline BRCA1-2 Pathogenic Variants. Cancer Med 2025; 14:e70364. [PMID: 39861955 PMCID: PMC11761426 DOI: 10.1002/cam4.70364] [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: 01/14/2024] [Revised: 04/07/2024] [Accepted: 10/13/2024] [Indexed: 01/27/2025] Open
Abstract
INTRODUCTION Pancreatic cancer arising in the context of BRCA predisposition may benefit from poly(ADP-ribose) polymerase inhibitors. We analyzed real-world data on the impact of olaparib on survival in metastatic pancreatic cancer patients harboring germline BRCA mutations in Italy, where olaparib is not reimbursed for this indication. METHODS Clinico/pathological data of pancreatic cancer patients with documented BRCA1-2 germline pathogenic variants who had received first-line chemotherapy for metastatic disease were collected from 23 Italian oncology departments and the impact of olaparib exposure on overall survival (OS) was analyzed. RESULTS Of 114, 53 BRCA-mutant pancreatic cancer patients had received olaparib for metastatic disease. OS was significantly longer in patients who were exposed to olaparib (hazard ratio [HR] 0.568, 95% confidence interval [CI] 0.351-0.918, log-rank p = 0.02) in any setting/line of treatment; similar results were obtained for patients who received olaparib as maintenance treatment (in any line of treatment), patients who had stage IV disease at diagnosis, and patients who did not experience progressive disease as their best response to first-line chemotherapy. Exposure to olaparib in the first-line maintenance setting after platinum-based chemotherapy, however, did not significantly impact survival. At multivariate analysis, CA19.9 levels at diagnosis and response to first-line chemotherapy were independently prognostic; however, when response to chemotherapy was excluded, any exposure to olaparib was a significant independent predictor of longer OS, together with CA19.9 levels. CONCLUSION The real-world data presented here support the use of olaparib for metastatic disease in germline BRCA-mutant pancreatic cancer patients, as it may significantly prolong survival.
Collapse
|
7
|
Lau-Min KS, Symecko H, Spielman K, Mann D, Hood R, Rathore S, Wolfe C, Gabriel PE, Rendle KA, Nathanson KL, Reiss KA, Domchek SM. Integration of Germline Genetic Testing Into Routine Clinical Practice for Patients With Pancreatic Adenocarcinoma. JCO Oncol Pract 2025; 21:170-177. [PMID: 39024535 PMCID: PMC11747919 DOI: 10.1200/op.24.00356] [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: 04/27/2024] [Revised: 05/31/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024] Open
Abstract
PURPOSE Germline genetic testing (GT) is recommended for all patients with pancreatic ductal adenocarcinoma (PDAC), but the traditional clinical genetics infrastructure is limited in addressing the unique needs of this population. We describe the integration of point of care (POC) GT into routine clinical practice for all patients with PDAC at an academic medical center. METHODS We developed a clinical POC workflow that leverages electronic health record (EHR) tools and behavioral nudges to enhance the sustainability and scalability of our previously described research-based POC model. For each of the research and clinical POC cohorts, we calculated the percentage of eligible patients who underwent GT. We used Wilcoxon rank-sum and Pearson's chi-squared tests to compare patients who did and did not undergo GT. We conducted surveys among oncology clinicians to evaluate the acceptability, appropriateness, and feasibility of the clinical POC model. RESULTS The research POC cohort included 905 patients, of whom 694 (76.7%) underwent GT. The clinical POC cohort included 148 patients, of whom 126 (85.1%) underwent GT. Patients who underwent GT in the research POC cohort were significantly younger (median age, 67.0 v 70.9 years; P = .031) and more likely to be White (82.1% v 68.7%; P < .001) and commercially insured (41.8% v 28.0%; P < .001) compared with those who did not; there were no significant differences between GT groups in the clinical POC cohort. Oncology clinicians found the clinical POC model to be acceptable (mean 4.4/5), appropriate (4.6/5), feasible (4.0/5), and have a positive impact on their patients (4.9/5). CONCLUSION A clinical POC model leveraging EHR tools and behavioral nudges is acceptable, appropriate, feasible, and associated with a >85% GT rate among patients with PDAC.
Collapse
Affiliation(s)
- Kelsey S. Lau-Min
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Heather Symecko
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelsey Spielman
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Derek Mann
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Hood
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Srishti Rathore
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Catherine Wolfe
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter E. Gabriel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katharine A. Rendle
- Department of Family Medicine and Community Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katherine L. Nathanson
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kim A. Reiss
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M. Domchek
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
8
|
Ekstrom TL, Rosok RM, Abdelrahman AM, Parassiadis C, Manjunath M, Dittrich MY, Wang X, Kutschat AP, Kanakan A, Rajput A, Schacherer N, Lukic T, Carlson DM, Thiel J, Kopp W, Stroebel P, Ellenrieder V, Gaedcke J, Dong M, Najafova Z, Truty MJ, Hessmann E, Johnsen SA. Glucocorticoid receptor suppresses GATA6-mediated RNA polymerase II pause release to modulate classical subtype identity in pancreatic cancer. Gut 2025:gutjnl-2024-334374. [PMID: 39884837 DOI: 10.1136/gutjnl-2024-334374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 12/23/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer with a 5-year survival rate of 12%. It has two major molecular subtypes: classical and basal, regulated by the master transcription factors (MTFs) GATA6 and ΔNp63, respectively. OBJECTIVE This study sought to uncover the transcriptional regulatory mechanisms controlling PDAC subtype identity. DESIGN We integrated primary tumour single-cell RNA-seq, patient-derived xenograft RNA-seq and multispectral imaging to identify MTF-dependent, subtype-specific markers. We created subtype-specific fluorescent reporter systems and conducted drug screenings to find actionable targets. We analysed chromatin accessibility (ATAC-seq), genome-wide occupancy (ChIP-seq) for epigenetic status (H3K27ac), MTFs (GATA6, ΔNp63), RNA polymerase II (Pol II), H3K4me3-anchored chromatin topology (HiChIP) and nascent RNA capture sequencing (PRO-seq). Additionally, we used nuclease-dead Cas9 (dCas9) to manipulate transcriptional regulatory mechanisms. RESULTS Our approach identified glucocorticoid receptor (GR) agonists as agents that suppress the classical transcriptional programme by interacting with GATA6. GATA6 regulates classical-specific transcription through promoter-proximal pause release. Depletion of GATA6 increased Pol II occupancy at GATA6-bound enhancers and transcriptional start sites, stabilising enhancer-promoter interactions. Artificially inducing pausing at GATA6-bound enhancers with dCas9 abrogated target gene expression and induced pausing at both the enhancer and target gene promoter. Conversely, in basal PDAC ΔNp63 promotes Pol II recruitment and stabilises enhancer-promoter interactions. CONCLUSION This study provides new insights into the transcriptional control and role of GR agonists in controlling PDAC molecular subtype identity.
Collapse
Affiliation(s)
- Thomas L Ekstrom
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | - Raya M Rosok
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | | | | | | | | | - Xin Wang
- Department of General, Visceral & Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Ana P Kutschat
- Department of General, Visceral & Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Akshay Kanakan
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Ashish Rajput
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | | | - Teodora Lukic
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Danielle M Carlson
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Julia Thiel
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Stuttgart, Germany
| | - Waltraut Kopp
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
- Clinical Research Group 5002, University Medical Center Göttingen, Göttingen, Germany
| | - Philipp Stroebel
- Clinical Research Group 5002, University Medical Center Göttingen, Göttingen, Germany
- Institute of Pathology, University Medical Center, Göttingen, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
- Clinical Research Group 5002, University Medical Center Göttingen, Göttingen, Germany
| | - Jochen Gaedcke
- Department of General, Visceral & Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
- Clinical Research Group 5002, University Medical Center Göttingen, Göttingen, Germany
- Department of General & Visceral Surgery, Karlsruhe Municipal Hospital, Karlsruhe, Germany
| | - Meng Dong
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Stuttgart, Germany
| | | | - Mark J Truty
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Elisabeth Hessmann
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
- Clinical Research Group 5002, University Medical Center Göttingen, Göttingen, Germany
| | - Steven A Johnsen
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Yousef M, Hurd MW, Yousef A, Ludmir EB, Pillai AB, Peterson J, Koay EJ, Albarouki S, Tzeng CW, Snyder R, Katz MHG, Wang H, Overman MJ, Maitra A, Pant S, Smaglo BG, Wolff RA, Yao J, Shen JP, Zhao D. Clinical and molecular characteristics of patients with brain metastasis secondary to pancreatic ductal adenocarcinoma. Oncologist 2025; 30:oyae182. [PMID: 39014543 PMCID: PMC11783327 DOI: 10.1093/oncolo/oyae182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/21/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND The prognosis for patients with pancreatic ductal adenocarcinoma (PDAC) is poor. Secondary brain metastasis (Br-M) occurs in less than 1% of patients. Clinical characteristics and molecular alterations have not been characterized in this rare patients' subset. MATERIALS AND METHODS The Foundry software platform was used to retrospectively query electronic health records for patients with Br-M secondary to PDAC from 2005 to 2023; clinical, molecular, and overall survival (OS) data were analyzed. RESULTS Br-M was diagnosed in 44 patients with PDAC. Median follow-up was 78 months; median OS from initial PDAC diagnosis was 47 months. Median duration from PDAC diagnosis to Br-M detection was 24 months; median OS from Br-M diagnosis was 3 months. At Br-M diagnosis, 82% (n = 36) of patients had elevated CA19-9. Lung was the most common preexisting metastatic location (71%) with Br-M, followed by liver (66%). Br-M were most frequently observed in the frontal lobe (34%, n = 15), cerebellar region (23%, n = 10), and leptomeninges (18%, n = 8). KRAS mutations were detected in 94.1% (n = 16) of patients who had molecular data available (n = 17) with KRASG12V being the most frequent subtype 47% (n = 8); KRASG12D in 29% (n = 5); KRASG12R in 18% (n = 3). Patients who underwent Br-M surgical resection (n = 5) had median OS of 8.6 months, while median OS following stereotactic radiosurgery only (n = 11) or whole-brain radiation only (n = 20) was 3.3 and 2.8 months, respectively. CONCLUSION Br-M is a late PDAC complication, resulting in an extremely poor prognosis especially in leptomeningeal disease. KRAS was mutated in 94.1% of the patients and the KRASG12V subtype was prevalent.
Collapse
Affiliation(s)
- Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mark W Hurd
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ethan B Ludmir
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ashwathy B Pillai
- Department of Hospital Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jennifer Peterson
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Eugene J Koay
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sali Albarouki
- Department of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States
| | - Ching-Wei Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rebecca Snyder
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Huamin Wang
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anirban Maitra
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Brandon G Smaglo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John P Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dan Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
10
|
Blanco Abad C, Gomila Pons P, Campos Ramírez S, Álvarez Alejandro M, Torres Ramón MI, Miramar Gallart MD, Izquierdo Álvarez S, Polo Marques E, Pazo Cid R. Hereditary Pancreatic Cancer: Advances in Genetic Testing, Early Detection Strategies, and Personalized Management. J Clin Med 2025; 14:367. [PMID: 39860372 PMCID: PMC11766428 DOI: 10.3390/jcm14020367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/29/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a five-year survival rate of approximately 13% for advanced stages. While the majority of PDAC cases are sporadic, a significant subset is attributable to hereditary and familial predispositions, accounting for approximately 25% of cases. This article synthesizes recent advancements in the understanding, detection, and management of hereditary pancreatic cancer (PC). Results: Our review highlights the critical role of genetic testing (GT) in identifying high-risk individuals (HRIs), with germline pathogenic variants (PVs) found in up to 20% of hereditary PDAC cases. Since the implementation of next-generation sequencing (NGS) panels in 2014, detection capabilities have been significantly enhanced. HRIs can be included in screening programs that facilitate the early detection of PDAC. Early detection strategies, including the use of microribonucleic acid (miRNAs) signatures and novel imaging techniques like hyperpolarized 13C-magnetic resonance spectroscopy (MRS) have shown promising results. The identification of germline pathogenic variants (PVs) or mutations in homologous recombination (HR) genes plays a predictive role in the response to various treatments, prolonging patient survival. Discussion: Universal germline testing for PDAC, as recommended by the National Comprehensive Cancer Network (NCCN), is now a standard practice, facilitating the identification of at-risk individuals and enabling targeted surveillance and intervention. Multidisciplinary management, integrating genetic counseling, imaging, and gastrointestinal services, is essential for optimizing outcomes. Conclusions: Advances in genetic testing and biomarker research are transforming the landscape of hereditary PC management. Early detection and personalized treatment strategies are pivotal in improving survival rates. Ongoing multi-institutional research efforts are crucial for validating biomarkers and developing preventive measures, ultimately aiming to reduce the burden of this aggressive cancer.
Collapse
Affiliation(s)
- Carmen Blanco Abad
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
| | - Paula Gomila Pons
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
| | - Sara Campos Ramírez
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
| | - María Álvarez Alejandro
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
- Medical Oncology Department, Hospital Clinico Universitario Lozano Blesa, 50009 Zaragoza, Spain
| | - María Irene Torres Ramón
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
- Medical Oncology Department, Hospital Clinico Universitario Lozano Blesa, 50009 Zaragoza, Spain
| | | | - Silvia Izquierdo Álvarez
- Genetics Unit, Biochemistry Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
| | - Eduardo Polo Marques
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
| | - Roberto Pazo Cid
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Medical Oncology Department, Hospital Clinico Universitario Lozano Blesa, 50009 Zaragoza, Spain
- Department of Medicine, Psychiatry and Dermatology, Faculty of Medicine, Zaragoza University, 50009 Zaragoza, Spain
| |
Collapse
|
11
|
Katona BW, Lubinski J, Pal T, Huzarski T, Foulkes WD, Moller P, Eisen A, Randall Armel S, Neuhausen SL, Raj R, Aeilts A, Singer CF, Bordeleau L, Karlan B, Olopade O, Tung N, Zakalik D, Kotsopoulos J, Fruscio R, Eng C, Sun P, Narod SA. The incidence of pancreatic cancer in women with a BRCA1 or BRCA2 mutation. Cancer 2025; 131:e35666. [PMID: 39611336 DOI: 10.1002/cncr.35666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/17/2024] [Accepted: 08/12/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND The lifetime risk of pancreatic cancer in women with a germline mutation in BRCA1 and BRCA2 is not well established. In an international prospective cohort of female carriers of BRCA1 and BRCA2 mutations, the cumulative incidence of pancreatic cancer from age 40 until 80 years was estimated. METHODS A total of 8295 women with a BRCA1 or BRCA2 mutation were followed for new cases of pancreatic cancer. Subjects were followed from the date of baseline questionnaire or age 40 years (whichever came last) until a new diagnosis of pancreatic cancer, death from another cause, or date of last follow-up. RESULTS Thirty-four incident pancreatic cancer cases were identified in the cohort. The annual risk of pancreatic cancer between age 40 and 80 years was 0.04% for BRCA1 carriers and 0.09% for BRCA2 carriers. Via the Kaplan-Meier method, the cumulative incidence from age 40 to 80 years was 2.2% (95% CI, 1.1%-4.3%) for BRCA1 carriers and 2.7% (95% CI, 1.3%-5.4%) for BRCA2 carriers. Only two of the 34 cases reported a first-degree relative with pancreatic cancer (hazard ratio, 4.75; 95% CI, 1.13-19.9; p = .03). Risk factors for pancreatic cancer included alcohol intake and a history of diabetes. The 5-year survival rate for the 34 cases was 8.8%. CONCLUSIONS The lifetime risk of pancreatic cancer is approximately 2% in women with a BRCA1 mutation and 3% for women with a BRCA2 mutation. The poor survival in hereditary pancreatic cancer underscores the need for novel antitumoral strategies.
Collapse
Affiliation(s)
- Bryson W Katona
- Basser Center for BRCA and Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jan Lubinski
- International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Tuya Pal
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tomasz Huzarski
- International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - William D Foulkes
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Pal Moller
- Institute of Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Andrea Eisen
- Sunnybrook Regional Cancer Centre, Toronto, Ontario, Canada
| | - Susan Randall Armel
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
- Division of Gynecologic Oncology, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Susan L Neuhausen
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Rebecca Raj
- Women's College Research Institute, Toronto, Ontario, Canada
| | - Amber Aeilts
- Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, Ohio, USA
| | - Christian F Singer
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Louise Bordeleau
- Department of Oncology, Juravinski Cancer Centre, Hamilton, Ontario, Canada
| | - Beth Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Olufunmilayo Olopade
- Department of Medicine and Human Genetics, University of Chicago, Chicago, Illinois, USA
| | - Nadine Tung
- Cancer Risk and Prevention Program, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Dana Zakalik
- Cancer Genetics Program, Beaumont Hospital, Royal Oak, Michigan, USA
| | - Joanne Kotsopoulos
- Women's College Research Institute, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Robert Fruscio
- Department of Medicine and Surgery, University of Milan Bicocca, IRCCS San Gerardo, Monza, Italy
| | - Charis Eng
- Center for Personalized Genetic Healthcare, Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ping Sun
- Women's College Research Institute, Toronto, Ontario, Canada
| | - Steven A Narod
- Women's College Research Institute, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
12
|
He X, Zhong L, Wang N, Zhao B, Wang Y, Wu X, Zheng C, Ruan Y, Hou J, Luo Y, Yin Y, He Y, Xiang AP, Wang J. Gastric Cancer Actively Remodels Mechanical Microenvironment to Promote Chemotherapy Resistance via MSCs-Mediated Mitochondrial Transfer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404994. [PMID: 39392399 DOI: 10.1002/advs.202404994] [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] [Received: 05/08/2024] [Revised: 09/03/2024] [Indexed: 10/12/2024]
Abstract
Chemotherapy resistance is the main reason of treatment failure in gastric cancer (GC). However, the mechanism of oxaliplatin (OXA) resistance remains unclear. Here, we demonstrate that extracellular mechanical signaling plays crucial roles in OXA resistance within GC. We selected OXA-resistant GC patients and analyzed tumor tissues by single-cell sequencing, and found that the mitochondrial content of GC cells increased in a biosynthesis-independent manner. Moreover, we found that the increased mitochondria of GC cells were mainly derived from mesenchymal stromal cells (MSCs), which could repair the mitochondrial function and reduce the levels of mitophagy in GC cells, thus leading to OXA resistance. Furthermore, we investigated the underlying mechanism and found that mitochondrial transfer was mediated by mechanical signals of the extracellular matrix (ECM). After OXA administration, GC cells actively secreted ECM in the tumor microenvironment (TEM), increasing matrix stiffness of the tumor tissues, which promoted mitochondria to transfer from MSCs to GC cells via microvesicles (MVs). Meanwhile, inhibiting the mechanical-related RhoA/ROCK1 pathway could alleviate OXA resistance in GC cells. In summary, these results indicate that matrix stiffness could be used as an indicator to identify chemotherapy resistance, and targeting mechanical-related pathway could effectively alleviate OXA resistance and improve therapeutic efficacy.
Collapse
Affiliation(s)
- Xin He
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Li Zhong
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Nan Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Baiwei Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yannan Wang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinxiang Wu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Changyu Zheng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Yueheng Ruan
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Jianfeng Hou
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yusheng Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yuehan Yin
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yulong He
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiancheng Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
13
|
Moretti M, Farina A, Angeloni A, Anastasi E. Emerging horizons on molecular and circulating biomarkers in pancreatic adenocarcinoma. Front Oncol 2024; 14:1483306. [PMID: 39575418 PMCID: PMC11578827 DOI: 10.3389/fonc.2024.1483306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most aggressive and invasive type of pancreatic cancer and is expected to soon become the second leading cause of cancer-associated death. The high mortality rate is due to the clinical features that allow asymptomatic progression to advanced stages, a period when current therapeutic treatments have limited efficacy. To address these challenges, researchers are focused on identifying new molecular and circulating markers for early PDAC detection and precision medicine. In this mini-review, we report the most well-known and recently identified molecular and circulating biomarkers. This study aimed to emphasize the need for continued innovative research to develop diagnostic algorithms and therapies to improve the management of patients with PDAC.
Collapse
Affiliation(s)
| | | | | | - Emanuela Anastasi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
14
|
Carconi C, Bosi C, Scartozzi M, Cergnul M, Cinausero M, Faloppi L, Garajova I, Lonardi S, Pecora I, Pisanu L, Spadi R, Spallanzani A, Peretti U, Macchini M, Orsi G, Reni M. A pilot study of chlorambucil in pre-treated metastatic pancreatic adenocarcinoma patients bearing germline BRCA or other DNA damage repair system variants. Pancreatology 2024; 24:1066-1072. [PMID: 39277480 DOI: 10.1016/j.pan.2024.09.006] [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: 04/16/2024] [Revised: 08/27/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
BACKGORUND Pancreatic adenocarcinoma remains a malignancy with a grim prognosis and scarce personalized treatment options. Pathogenic variants of DNA damage repair (DDR) genes are emerging as molecular targets, as they confer a higher sensitivity to DNA-damaging agents. This study aimed at assessing the activity of chlorambucil as salvage therapy in metastatic pancreatic cancer patients bearing a germline pathogenetic variant or variant of uncertain significance on a DDR-related gene. METHODS Platinum-pretreated metastatic pancreatic cancer patients harbouring a germline variant on a DDR gene received chlorambucil at a daily oral dose of 6 mg/m2 for 42 every 56 days for the first cycle and for 14 every 28 days for the following cycles, until disease progression or unacceptable toxicity. The primary endpoint was 6-month progression-free survival rate (PFS-6). Median progression-free survival (PFS) and overall survival (OS) were secondarily described. RESULTS Twenty patients were enrolled between December 2020 and September 2022. PFS-6 was 5%, median PFS and OS were 1.6 months and 3.0 months, respectively. Grade-3 adverse events were observed in 25% of patients, while no Grade-4 toxicity was reported. CONCLUSIONS Single agent chlorambucil did not show sufficient signal of activity to warrant its further investigation in metastatic pancreatic cancer patients bearing a DDR-related germline alteration.
Collapse
Affiliation(s)
- Catia Carconi
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Carlo Bosi
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Mario Scartozzi
- Medical Oncology, University and University Hospital, Cagliari, Italy
| | - Massimiliano Cergnul
- Medical Oncology, Ospedale Civile di Legnano - ASST Ovest Milanese, Legnano, Italy
| | - Marika Cinausero
- Department of Oncology, Azienda Sanitaria Universitaria Friuli Centrale Santa Maria Della Misericordia, Udine, Italy
| | - Luca Faloppi
- Medical Oncology Unit, Ospedali Santa Maria Della Pietà e Bartolomeo Eustachio - AST di Macerata, Camerino, San Severino Marche, Italy
| | - Ingrid Garajova
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Irene Pecora
- Department of Medical Oncology, Azienda Toscana Sud-Est, Misericordia Hospital, Grosseto, Italy
| | | | - Rosella Spadi
- Department of Oncology, Medical Oncology, 1, Città Della Salute e Della Scienza, Turin, Italy
| | - Andrea Spallanzani
- Division of Oncology, Department of Oncology and Hematology, University Hospital Modena, Modena, Italy
| | - Umberto Peretti
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Marina Macchini
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Giulia Orsi
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Michele Reni
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
15
|
Espona-Fiedler M, Patthey C, Lindblad S, Sarró I, Öhlund D. Overcoming therapy resistance in pancreatic cancer: New insights and future directions. Biochem Pharmacol 2024; 229:116492. [PMID: 39153553 DOI: 10.1016/j.bcp.2024.116492] [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: 04/05/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Pancreatic adenocarcinoma (PDAC) is predicted to become the second leading cause of cancer deaths by 2030 and this is mostly due to therapy failure. Limited treatment options and resistance to standard-of-care (SoC) therapies makes PDAC one of the cancer types with poorest prognosis and survival rates [1,2]. Pancreatic tumors are renowned for their poor response to therapeutic interventions including targeted therapies, chemotherapy and radiotherapy. Herein, we review hallmarks of therapy resistance in PDAC and current strategies aiming to tackle escape mechanisms and to re-sensitize cancer cells to therapy. We will further provide insights on recent advances in the field of drug discovery, nanomedicine, and disease models that are setting the ground for future research.
Collapse
Affiliation(s)
- Margarita Espona-Fiedler
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden.
| | - Cedric Patthey
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden
| | - Stina Lindblad
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden
| | - Irina Sarró
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Universitat de Barcelona, Barcelona, Spain
| | - Daniel Öhlund
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden.
| |
Collapse
|
16
|
Okano N, Kawai M, Ueno M, Yu X, Inoue Y, Takahashi S, Wang W, Takahashi H, Okamura Y, Morinaga S, Matsumoto I, Shimizu Y, Yoshida K, Yamamoto T, Ohtsuka M, Inokawa Y, Nara S, Tamura J, Shinoda S, Yamamoto K, Yamaue H, Furuse J. Outcomes of patients with initially unresectable pancreatic cancer who underwent conversion surgery after FOLFIRINOX or gemcitabine plus nab-paclitaxel chemotherapy: A multicenter retrospective cohort study (PC-CURE-1). JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:816-829. [PMID: 39150050 PMCID: PMC11589395 DOI: 10.1002/jhbp.12066] [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: 08/17/2024]
Abstract
BACKGROUND The efficacy and safety of conversion surgery (CS) after FOLFIRINOX or gemcitabine plus nab-paclitaxel (GnP) chemotherapy in patients with initially unresectable pancreatic cancer (PC) remains unclear. METHODS This multicenter retrospective cohort study enrolled patients, between 2014 and 2018, with initially locally advanced or metastatic PC who were considered candidates for CS following FOLFIRINOX or GnP chemotherapy. They were classified into surgery (207 patients [194 resection and 13 exploratory laparotomy only]) and continued chemotherapy (10 patients, control) groups. The primary endpoint was overall survival (OS) from the day of diagnosis of potentially curative resection on imaging studies, with an expected hazard ratio (HR) of 0.7. RESULTS OS in the surgery group was longer than that in the control group (HR, 0.47; 95% confidence interval [CI]: 0.24-0.93). The median OS was 34.4 (95% CI: 27.9-43.4) and 19.8 (95% CI: 14.9-31.1) months in the surgery and control groups, respectively. The Clavien-Dindo grade ≥ IIIa postoperative complication and in-hospital mortality rates were 19.6% and 0.5%, respectively. Multivariate analysis revealed that preoperative chemotherapy duration was not associated with OS. CONCLUSIONS CS, following a favorable response to FOLFIRINOX or GnP chemotherapy, improved initially unresectable PC prognosis (specifically, OS), regardless of the chemotherapy duration.
Collapse
Affiliation(s)
- Naohiro Okano
- Department of Medical OncologyKyorin University Faculty of MedicineTokyoJapan
| | - Manabu Kawai
- Second Department of SurgeryWakayama Medical University School of MedicineWakayamaJapan
| | - Makoto Ueno
- Department of GastroenterologyKanagawa Cancer CenterYokohamaJapan
| | - Xianjun Yu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic SurgeryCancer Institute Hospital, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Shinichiro Takahashi
- Department of Hepatobiliary and Pancreatic SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Wenquan Wang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
| | - Hidenori Takahashi
- Department of Gastroenterological SurgeryOsaka International Cancer InstituteOsakaJapan
| | - Yukiyasu Okamura
- Division of Hepato‐Biliary‐Pancreatic SurgeryShizuoka Cancer Center HospitalShizuokaJapan
| | - Soichiro Morinaga
- Department of Hepato‐Biliary and Pancreatic SurgeryKanagawa Cancer CenterYokohamaJapan
| | - Ippei Matsumoto
- Department of SurgeryKindai University Faculty of MedicineOsakasayamaJapan
| | - Yasuhiro Shimizu
- Department of Gastroenterological SurgeryAichi Cancer Center HospitalNagoyaJapan
| | - Kazuhiro Yoshida
- Department of Surgical OncologyGifu University Graduate School of MedicineGifuJapan
| | | | - Masayuki Ohtsuka
- Department of General Surgery, Graduate School of MedicineChiba UniversityChibaJapan
| | - Yoshikuni Inokawa
- Department of Gastroenterological SurgeryNagoya University Graduate School of MedicineNagoyaJapan
| | - Satoshi Nara
- Department of Hepatobiliary and Pancreatic SurgeryNational Cancer Center HospitalTokyoJapan
| | - Jun Tamura
- Department of BiostatisticsYokohama City University School of MedicineYokohamaJapan
| | - Satoru Shinoda
- Department of BiostatisticsYokohama City University School of MedicineYokohamaJapan
| | - Kouji Yamamoto
- Department of BiostatisticsYokohama City University School of MedicineYokohamaJapan
| | - Hiroki Yamaue
- Second Department of SurgeryWakayama Medical University School of MedicineWakayamaJapan
| | - Junji Furuse
- Department of Medical OncologyKyorin University Faculty of MedicineTokyoJapan
- Department of GastroenterologyKanagawa Cancer CenterYokohamaJapan
| | | |
Collapse
|
17
|
Archasappawat S, Al-Musawi F, Liu P, Lee E, Hwang CI. Familial Pancreatic Cancer Research: Bridging Gaps in Basic Research and Clinical Application. Biomolecules 2024; 14:1381. [PMID: 39595558 PMCID: PMC11592027 DOI: 10.3390/biom14111381] [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: 08/26/2024] [Revised: 10/07/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Familial pancreatic cancer (FPC) represents a significant yet underexplored area in pancreatic cancer research. Basic research efforts are notably limited, and when present, they are predominantly centered on the BRCA1 and BRCA2 mutations due to the scarcity of other genetic variants associated with FPC, leading to a limited understanding of the broader genetic landscape of FPC. This review examines the current state of FPC research, focusing on the molecular mechanisms driving pancreatic ductal adenocarcinoma (PDAC) progression. It highlights the role of homologous recombination (HR) and its therapeutic exploitation via synthetic lethality with PARP inhibitors in BRCA1/2-deficient tumors. The review discusses various pre-clinical models of FPC, including conventional two-dimensional (2D) cell lines, patient-derived organoids (PDOs), patient-derived xenografts (PDXs), and genetically engineered mouse models (GEMMs), as well as new advancements in FPC research.
Collapse
Affiliation(s)
- Suyakarn Archasappawat
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
- University of California Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - Fatimah Al-Musawi
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - Peiyi Liu
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - EunJung Lee
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - Chang-il Hwang
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
- University of California Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
18
|
Sudo K, Nakamura Y, Ueno M, Furukawa M, Mizuno N, Kawamoto Y, Okano N, Umemoto K, Asagi A, Ozaka M, Ohtsubo K, Shimizu S, Matsuhashi N, Itoh S, Matsumoto T, Satoh T, Okuyama H, Goto M, Hasegawa H, Yamamoto Y, Odegaard JI, Bando H, Yoshino T, Ikeda M, Morizane C. Clinical utility of BRCA and ATM mutation status in circulating tumour DNA for treatment selection in advanced pancreatic cancer. Br J Cancer 2024; 131:1237-1245. [PMID: 39198618 PMCID: PMC11443054 DOI: 10.1038/s41416-024-02834-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Identification of homologous recombination deficiency (HRD) remains a challenge in advanced pancreatic cancer (APC). We investigated the utility of circulating tumour DNA (ctDNA) profiling in the assessment of BRCA1/2 and ATM mutation status and treatment selection in APC. METHODS We analysed clinical and ctDNA data of 702 patients with APC enroled in GOZILA, a ctDNA profiling study using Guardant360. RESULTS Inactivating BRCA1/2 and ATM mutations were detected in 4.8% (putative germline, 3.7%) and 4.4% (putative germline, 0.9%) of patients, respectively. Objective response (63.2% vs. 16.2%) and PFS (HR 0.55, 95% CI 0.32-0.93) on platinum-containing chemotherapy were significantly better in patients with putative germline BRCA1/2 (gBRCA) mutation than those without. In contrast, putative gBRCA mutation had no impact on the efficacy of gemcitabine plus nab-paclitaxel. In 2 patients treated with platinum-containing therapy, putative BRCA2 reversion mutations were detected. Three of seven patients with somatic BRCA mutations responded to platinum-containing therapy, while only one of four with putative germline ATM mutations did. One-third of somatic ATM mutations were in genomic loci associated with clonal haematopoiesis. CONCLUSION Comprehensive ctDNA profiling provides clinically relevant information regarding HRD status. It can be a practical, convenient option for HRD screening in APC.
Collapse
Affiliation(s)
- Kentaro Sudo
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
- Translational Research Support Office, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Masayuki Furukawa
- Department of Hepato-Biliary-Pancreatology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Nobumasa Mizuno
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yasuyuki Kawamoto
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Mitaka, Japan
| | - Kumiko Umemoto
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Akinori Asagi
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Koushiro Ohtsubo
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Satoshi Shimizu
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Nobuhisa Matsuhashi
- Department of Gastroenterological Surgery and Pediatric Surgery, Gifu University, Graduate School of Medicine, Gifu, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, Japan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshihiko Matsumoto
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Suita, Japan
| | - Hiroyuki Okuyama
- Department of Medical Oncology, Kagawa University Hospital, Kagawa, Japan
| | - Masahiro Goto
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University Hospital, Osaka, Japan
| | - Hiroko Hasegawa
- Department of Gastroenterology and Hepatology, NHO, Osaka National Hospital, Osaka, Japan
| | - Yoshiyuki Yamamoto
- Department of Gastroenterology, University of Tsukuba Hospital, Tsukuba, Japan
| | | | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, Department for the Promotion of Drug and Diagnostic Development, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
19
|
Hanaoka T, Okuwaki K, Nakamura K, Okada S, Nishizawa N, Watanabe M, Iwai T, Adachi K, Kumamoto Y, Kusano C. High likelihood of BRCA2 reversion mutation in pancreatic cancer post-platinum-based chemotherapy: a case study. Int Cancer Conf J 2024; 13:493-498. [PMID: 39398920 PMCID: PMC11464854 DOI: 10.1007/s13691-024-00715-0] [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: 06/23/2024] [Accepted: 08/11/2024] [Indexed: 10/15/2024] Open
Abstract
A 54-year-old man with resectable pancreatic cancer and abnormally high levels of carbohydrate antigen 19-9 (CA19-9) underwent 6 months of platinum-based chemotherapy. This treatment substantially reduced the primary tumor size and normalized CA19-9 levels. Subsequently, radical surgery was conducted. However, eight months post-surgery, CA19-9 levels re-elevated, and lymph-node recurrence was observed. The patient underwent treatment with poly(adenosine diphosphate ribose) polymerase inhibitors (PARPi) following the detection of frameshift L1904fs*5 via BRACAnalysis CDx. This mutation revealed a stop codon, leading to the inactivation of the BRCA function. Additionally, the patient tested positive for a mutation in the breast cancer susceptibility gene 2 (BRCA2). Two months after starting PARPi, there was evidence of tumor shrinkage. Nevertheless, 5 months later, CA19-9 levels increased again, and new metastatic tumors in the liver were identified. Genomic profiling test (FoundationOne CDx) of surgically resected specimens revealed a BRCA2 pL1908fs*2 mutation, indicating its location in the cis position on the same allele as the germline BRCA2 mutation. The pL1908fs*2 deletion, alongside the original L1904fs*5, resulted in three deletions, equating to one amino acid deletion. This deletion ultimately reversed the stop codon, leading to the restoration of BRCA2 functionality. Despite treatment with PARPi for postoperative recurrence, a sustained response was not achieved owing to BRCA reversion mutations. It is essential to acknowledge the rarity of BRCA reversion mutations, which limit the effectiveness of PARPi.
Collapse
Affiliation(s)
- Taro Hanaoka
- Department of Gastroenterology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374 Japan
| | - Kosuke Okuwaki
- Department of Gastroenterology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374 Japan
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582 Japan
| | - Shunji Okada
- Department of Gastroenterology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374 Japan
| | - Nobuyuki Nishizawa
- Department of General-Pediatric-Hepatobiliary Pancreatic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374 Japan
| | - Masafumi Watanabe
- Department of Gastroenterology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374 Japan
| | - Tomohisa Iwai
- Department of Gastroenterology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374 Japan
| | - Kai Adachi
- Department of Gastroenterology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374 Japan
| | - Yusuke Kumamoto
- Department of General-Pediatric-Hepatobiliary Pancreatic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374 Japan
| | - Chika Kusano
- Department of Gastroenterology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374 Japan
| |
Collapse
|
20
|
Crapé L, Geboes K, Kortbeek K, Naert E, Hoorens A, Berrevoet F, Van Heddeghem N, Ribeiro S. Long-term complete remission of two patients with synchronous liver metastasis from pancreatic cancer and underlying BRCA-2 mutation. Acta Gastroenterol Belg 2024; 87:521-523. [PMID: 39745039 DOI: 10.51821/87.4.11787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) has a known poor prognosis. For a select group, those with BRCA mutations, frontline platinum-based therapy and poly (ADPribose) polymerase inhibitors are options that can potentially lead to survival benefit. Patients and methods We present 2 cases of patients with BRCAmutated pancreatic cancer with liver metastases that achieved a remarkable long-term complete remission on platinum-based chemotherapy. Conclusion Germline testing for BRCA is important in PDAC because it influences treatment choices that impact survival. Complete responses with chemotherapy alone are rarely observed in metastatic PDAC, but may be seen upon treatment with platinum-based therapy.
Collapse
Affiliation(s)
- L Crapé
- Department of Gastroenterology, UZ Ghent, Ghent, Belgium
| | - K Geboes
- Department of Gastroenterology, UZ Ghent, Ghent, Belgium
| | - K Kortbeek
- Department of Gastroenterology, UZ Brussels, Brussels, Belgium
| | - E Naert
- Department of Gastroenterology, UZ Ghent, Ghent, Belgium
| | - A Hoorens
- Department of Gastroenterology, UZ Ghent, Ghent, Belgium
| | - F Berrevoet
- Department of Gastroenterology, UZ Ghent, Ghent, Belgium
| | | | - S Ribeiro
- Department of Gastroenterology, UZ Ghent, Ghent, Belgium
| |
Collapse
|
21
|
Li B, Zhang Q, Castaneda C, Cook S. Targeted Therapies in Pancreatic Cancer: A New Era of Precision Medicine. Biomedicines 2024; 12:2175. [PMID: 39457488 PMCID: PMC11505516 DOI: 10.3390/biomedicines12102175] [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: 08/16/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a leading cause of cancer mortality in the United States, presents significant treatment challenges due to its late diagnosis and poor prognosis. Despite advances, the five-year survival rates remain dismally low, with only a fraction of patients eligible for potentially curative surgical interventions. This review aims to comprehensively examine the current landscape of targeted therapies in PDAC, focusing on recent developments in precision medicine approaches. We explore various molecular targets, including KRAS mutations, DNA damage repair deficiencies, mismatch repair pathway alterations, and rare genetic fusions. The review discusses emerging therapies, such as PARP inhibitors, immune checkpoint inhibitors, and novel targeted agents, like RET and NTRK inhibitors. We analyze the results of key clinical trials and highlight the potential of these targeted approaches in specific patient subgroups. Recent developments in PDAC research have emphasized precision oncology, facilitated by next-generation sequencing and the identification of genetic and epigenetic alterations. This approach tailors treatments to individual genetic profiles, improving outcomes and reducing side effects. Significant strides have been made in classifying PDAC into various subtypes, enhancing therapeutic precision. The identification of specific mutations in genes like KRAS, along with advancements in targeted therapies, including small molecule inhibitors, offers new hope. Furthermore, emerging therapies targeting DNA repair pathways and immunotherapeutic strategies also show promising results. As research evolves, integrating these targeted therapies with conventional treatments might improve survival rates and quality of life for PDAC patients, underscoring the shift towards a more personalized treatment paradigm.
Collapse
Affiliation(s)
- Bingyu Li
- University of Wisconsin Hospitals and Clinics, Madison, WI 53792-2460, USA; (B.L.); (Q.Z.); (C.C.)
- School of Medicine, Tongji University, Shanghai 200092, China
| | - Qiong Zhang
- University of Wisconsin Hospitals and Clinics, Madison, WI 53792-2460, USA; (B.L.); (Q.Z.); (C.C.)
| | - Claire Castaneda
- University of Wisconsin Hospitals and Clinics, Madison, WI 53792-2460, USA; (B.L.); (Q.Z.); (C.C.)
| | - Shelly Cook
- University of Wisconsin Hospitals and Clinics, Madison, WI 53792-2460, USA; (B.L.); (Q.Z.); (C.C.)
| |
Collapse
|
22
|
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
|
23
|
Singh H, Nipp RD. Bridging the Divide: From Universal Germline Testing Guidance to Real-World Implementation in Pancreatic Cancer Care. JCO Oncol Pract 2024; 20:1012-1015. [PMID: 38718312 DOI: 10.1200/op.24.00237] [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: 03/18/2024] [Accepted: 04/04/2024] [Indexed: 08/15/2024] Open
Abstract
In this editorial accompanying manuscript the by Klatte and colleagues, Drs Singh, and Nipp review the data behind universal germline testing in pancreatic adenocarcinoma, and review possible reasons for and solutions continued inadequate rates of germline testing in our community. Making germline testing for all patients with pancreatic adenocarcinoma is critical and will require cross-disciplinary collaboration and innovation.
Collapse
Affiliation(s)
- Harshabad Singh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Ryan D Nipp
- University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Oklahoma City, OK
| |
Collapse
|
24
|
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
|
25
|
Rosso C, Marciano ND, Nathan D, Chen WP, McLaren CE, Osann KE, Flodman PL, Cho MT, Lee FC, Dayyani F, Zell JA, Valerin JB. Hereditary Cancer Clinics Improve Adherence to NCCN Germline Testing Guidelines for Pancreatic Cancer. J Natl Compr Canc Netw 2024; 22:299-305. [PMID: 38889755 PMCID: PMC11462954 DOI: 10.6004/jnccn.2023.7333] [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: 06/07/2023] [Accepted: 12/21/2023] [Indexed: 06/20/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, with a 5-year overall survival rate of 10%. In November 2018, NCCN recommended that all patients with PDAC receive genetic counseling (GC) and germline testing regardless of family history. We hypothesized that patients with PDAC were more likely to be referred for testing after this change to the guidelines, regardless of presumed predictive factors, and that compliance would be further improved following the implementation of a hereditary cancer clinic (HCC). METHODS We conducted a single-institution retrospective analysis of patients diagnosed with PDAC from June 2017 through December 2021 at University of California, Irvine. We compared rates of genetics referral among patients in different diagnostic eras: the 18-month period before the NCCN Guideline change (pre-NCCN era: June 2017 through November 2018), 14 months following the change (post-NCCN era: December 2018 through January 2020), and 18 months after the creation of an HCC (HCC era: June 2020 through December 2021). Family and personal cancer history, genetics referral patterns, and results of GC were recorded. Data were compared using chi-square, Fisher exact, and multivariate analyses. RESULTS A total of 335 patients were treated for PDAC (123 pre-NCCN, 109 post-NCCN, and 103 HCC) at University of California, Irvine. Demographics across groups were comparable. Prior to the guideline changes, 30% were referred to GC compared with 54.7% in the post-NCCN era. After the implementation of the HCC, 77.4% were referred to GC (P<.0001). The odds ratio (OR) for referral to GC among patients with a positive family history of cancer progressively decreased following the change (pre-NCCN era: OR, 11.90 [95% CI, 3.00-80.14]; post-NCCN era: OR, 3.39 [95% CI, 1.13-10.76]; HCC era: OR, 3.11 [95% CI, 0.95-10.16]). CONCLUSIONS The 2018 updates to the NCCN Guidelines for PDAC recommending germline testing for all patients with PDAC significantly increased GC referral rates at our academic medical center. Implementation of an HCC further boosted compliance with guidelines.
Collapse
Affiliation(s)
- Claudia Rosso
- School of Medicine, University of California, Irvine, Irvine, CA
| | | | - Deepika Nathan
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, Irvine, CA
| | - Wen-Pin Chen
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
| | - Christine E. McLaren
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
| | - Kathryn E. Osann
- School of Medicine, University of California, Irvine, Irvine, CA
| | - Pamela L. Flodman
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, Irvine, CA
| | - May T. Cho
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
- Division of Hematology/Oncology, Department of Medicine, UCI School of Medicine, Irvine, CA
| | - Fa-Chyi Lee
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
- Division of Hematology/Oncology, Department of Medicine, UCI School of Medicine, Irvine, CA
| | - Farshid Dayyani
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
- Division of Hematology/Oncology, Department of Medicine, UCI School of Medicine, Irvine, CA
| | - Jason A. Zell
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
- Division of Hematology/Oncology, Department of Medicine, UCI School of Medicine, Irvine, CA
| | - Jennifer B. Valerin
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
- Division of Hematology/Oncology, Department of Medicine, UCI School of Medicine, Irvine, CA
| |
Collapse
|
26
|
Yu KH. Advances in Systemic Therapy in Pancreatic Cancer. Hematol Oncol Clin North Am 2024; 38:617-627. [PMID: 38575456 DOI: 10.1016/j.hoc.2024.03.002] [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: 04/06/2024]
Abstract
Substantial progress has been made toward understanding biology and developing new therapies for pancreatic ductal adenocarcinoma (PDAC). In this review, new insights from genomic profiling, as well as implications for treatment and prognosis, are discussed. New standards of care approaches with a focus on drug therapies are discussed for the treatment of resectable and advanced PDAC. The role of targeted and immune therapies remains limited; cohorts likely to benefit from these approaches are discussed. Promising, preliminary results regarding experimental therapies are reviewed.
Collapse
Affiliation(s)
- Kenneth H Yu
- Gastrointestinal Oncology Service, Cell Therapy Service, Memorial Sloan Kettering Cancer Center, 300 E 66th Street, New York, NY 10065, USA.
| |
Collapse
|
27
|
Muzzolini M, Lupinacci R, Bachet JB, Lassoued D, Sauvanet A, Gaujoux S. Should liver metastases of pancreatic adenocarcinoma be resected? J Visc Surg 2024; 161:129-140. [PMID: 38262871 DOI: 10.1016/j.jviscsurg.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Affiliation(s)
- Milena Muzzolini
- Department of hepato-biliary, pancreatic surgery and liver transplantation, hôpital la Pitié-Salpêtrière, AP-HP, Paris, France; Université de Paris Cité, Paris, France.
| | - Renato Lupinacci
- UFR des sciences de la santé Simone-Veil, université Versailles Saint-Quentin en Yvelines/Paris Saclay, Montigny-le-Bretonneux, France; Digestive and oncological surgery department, université Paris Saclay, hôpital Ambroise-Paré. AP-HP, Boulogne-Billancourt, France
| | - Jean-Baptiste Bachet
- Sorbonne université, Paris, France; Oncology department, hôpital la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Donia Lassoued
- Oncology department, hôpital la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Alain Sauvanet
- Université de Paris Cité, Paris, France; Department of hepato-biliary, pancreatic surgery and liver transplantation, hôpital Beaujon, AP-HP, Clichy, France
| | - Sébastien Gaujoux
- Department of hepato-biliary, pancreatic surgery and liver transplantation, hôpital la Pitié-Salpêtrière, AP-HP, Paris, France; Sorbonne université, Paris, France
| |
Collapse
|
28
|
Koukaki T, Balgkouranidou I, Biziota E, Karayiannakis A, Bolanaki H, Karamitrousis E, Zarogoulidis P, Deftereos S, Charalampidis C, Ioannidis A, Matthaios D, Amarantidis K, Kakolyris S. Prognostic significance of BRCA1 and BRCA2 methylation status in circulating cell-free DNA of Pancreatic Cancer patients. J Cancer 2024; 15:2573-2579. [PMID: 38577595 PMCID: PMC10988318 DOI: 10.7150/jca.93184] [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: 12/13/2023] [Accepted: 02/08/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction: Pancreatic cancer is the most fatal cancer type in the world. Its high mortality is mostly correlated to the absence of symptoms and the difficulty in early diagnosis, which in the majority of the cases occurs when the disease has already spread metastasis. Nowadays, tests that could predict early diagnosis are not available yet and the number of prognostic tests is limited. Hence, there is an urgent need for biomarkers capable of detecting early development or the rapid progression of the disease. Patients and Methods: DNA methylation represents the most frequent epigenetic event among tumor suppressor genes that are involved in various carcinogenic pathways. In the recent study we have tried to evaluate, for the first time, the prognostic value of BRCA1 and BRCA2 methylation in the cell-free DNA of pancreatic cancer patients. Using methylation-specific real-time PCR we examined the methylation status of BRCA1 and BRCA2 in 55 patients with operable and 50 patients with metastatic pancreatic cancer. In the operable disease setting, BRCA1 was found to be methylated in 33/55 (63.5%) patients examined while BRCA2 was also highly methylated in 31/55 (56.3%). In the metastatic disease, BRCA1 was found to be methylated in 26/50 (52%) while BRCA2 was found methylated in 23/50 (46%). Results: All control samples were negative for BRCA1 orBRCA2 promoter methylation. Patients with operable pancreatic cancer and a methylated BRCA1 and BRCA2 promoter status had a statistically significant poorer outcome as compared with patients with a non-methylated one (p=0.012 and p=0.001, respectively). Conclusion: In this study plasma methylation of BRCA1 and BRCA2 represents a frequent event in both the operable as well as in the metastatic setting. BRCA1 and BRCA2 methylation was significant and correlated with decreased survival in patients with operable pancreatic cancer. A larger cohort of patients is required to further explore the potential of these findings as well as to investigate whether BRCA1/2 methylation in plasma could serve as a potential prognostic biomarker in pancreatic cancer.
Collapse
Affiliation(s)
- Triantafyllia Koukaki
- Department of Medical Oncology, Medical School, Democritus University of Thrace, Greece
| | - Ioanna Balgkouranidou
- Department of Medical Oncology, Medical School, Democritus University of Thrace, Greece
| | - Eirini Biziota
- Department of Medical Oncology, Medical School, Democritus University of Thrace, Greece
| | | | - Helen Bolanaki
- Department of 2nd Surgery, Medical School, Democritus University of Thrace, Greece
| | - Evangelos Karamitrousis
- University Medical Oncology department, Aristotle University of Thessaloniki, Papageorgiou General Hospital
| | - Paul Zarogoulidis
- Pulmonary Oncology Department, General Clinic Euromedica, Thessaloniki, Greece
| | - Savas Deftereos
- Radiology Department, Medical School, Democritus University of Thrace, Greece
| | | | - Aris Ioannidis
- Surgery Department, Genesis Private Clinic, Thessaloniki, Greece
| | | | - Kyriakos Amarantidis
- Department of Medical Oncology, Medical School, Democritus University of Thrace, Greece
| | - Stylianos Kakolyris
- Department of Medical Oncology, Medical School, Democritus University of Thrace, Greece
| |
Collapse
|
29
|
Ikezawa K, Urabe M, Kai Y, Takada R, Akita H, Nagata S, Ohkawa K. Comprehensive review of pancreatic acinar cell carcinoma: epidemiology, diagnosis, molecular features and treatment. Jpn J Clin Oncol 2024; 54:271-281. [PMID: 38109477 PMCID: PMC10925851 DOI: 10.1093/jjco/hyad176] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023] Open
Abstract
Pancreatic acinar cell carcinoma is a rare form (0.2-4.3%) of pancreatic neoplasm with unique clinical and molecular characteristics, which largely differ from pancreatic ductal adenocarcinoma. Pancreatic acinar cell carcinoma occurs more frequently in males and can occur in children. Serum lipase is elevated in 24-58% of patients with pancreatic acinar cell carcinoma. Pancreatic acinar cell carcinomas tend to be large at diagnosis (median tumour size: ~5 cm) and are frequently located in the pancreas head. Radiologically, pancreatic acinar cell carcinoma generally exhibits a solid appearance; however, necrosis, cystic changes and intratumoral haemorrhage can occur in larger lesions. Immunostaining is essential for the definitive diagnosis of pancreatic acinar cell carcinoma. Compared with pancreatic ductal adenocarcinoma, pancreatic acinar cell carcinoma has a more favourable prognosis. Although radical surgery is recommended for patients with pancreatic acinar cell carcinoma who do not have distant metastases, the recurrence rate is high. The effectiveness of adjuvant therapy for pancreatic acinar cell carcinoma is unclear. The response to FOLFIRINOX is generally favourable, and some patients achieve a complete response. Pancreatic acinar cell carcinoma has a different genomic profile compared with pancreatic ductal adenocarcinoma. Although genomic analyses have shown that pancreatic acinar cell carcinoma rarely has KRAS, TP53 and CDKN2A mutations, it has a higher prevalence of homologous recombination-related genes, including BRCA1/2 and ATM, than pancreatic ductal adenocarcinoma, suggesting high sensitivity to platinum-containing regimens and PARP inhibitors. Targeted therapies for genomic alternations are beneficial. Therefore, genetic testing is important for patients with pancreatic acinar cell carcinoma to choose the optimal therapeutic strategy.
Collapse
Affiliation(s)
- Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Makiko Urabe
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Yugo Kai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Ryoji Takada
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Hirofumi Akita
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Shigenori Nagata
- Department of Diagnostic Pathology and Cytology, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
30
|
Szentmartoni G, Mühl D, Csanda R, Szasz AM, Herold Z, Dank M. Predictive Value and Therapeutic Significance of Somatic BRCA Mutation in Solid Tumors. Biomedicines 2024; 12:593. [PMID: 38540206 PMCID: PMC10967875 DOI: 10.3390/biomedicines12030593] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 01/11/2025] Open
Abstract
Ten percent of patients with breast cancer, and probably somewhat more in patients with ovarian cancer, have inherited germline DNA mutations in the breast and ovarian cancer genes BRCA1 and BRCA2. In the remaining cases, the disease is caused by acquired somatic genetic and epigenetic alterations. Targeted therapeutic agents, such as poly ADP-ribose polymerases (PARP) inhibitors (PARPi), have emerged in treating cancers associated with germline BRCA mutations since 2014. The first PARPi was FDA-approved initially for ovarian cancer patients with germline BRCA mutations. Deleterious variants in the BRCA1/BRCA2 genes and homologous recombination deficiency status have been strong predictors of response to PARPi in a few solid tumors since then. However, the relevance of somatic BRCA mutations is less clear. Somatic BRCA-mutated tumors might also respond to this new class of therapeutics. Although the related literature is often controversial, recently published case reports and/or randomized studies demonstrated the effectiveness of PARPi in treating patients with somatic BRCA mutations. The aim of this review is to summarize the predictive role of somatic BRCA mutations and to provide further assistance for clinicians with the identification of patients who could potentially benefit from PARPi.
Collapse
Affiliation(s)
- Gyongyver Szentmartoni
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary
| | | | | | | | | | | |
Collapse
|
31
|
Ahn ER, Rothe M, Mangat PK, Garrett-Mayer E, Calfa CJ, Alva AS, Suhag V, Alese OB, Dotan E, Hamid O, Yang ES, Marr AS, Palmer MC, Thompson FL, Yost KJ, Gregory A, Grantham GN, Hinshaw DC, Halabi S, Schilsky RL. Olaparib in Patients With Pancreatic Cancer With BRCA1/ 2 Mutations: Results From the Targeted Agent and Profiling Utilization Registry Study. JCO Precis Oncol 2024; 8:e2300240. [PMCID: PMC10896473 DOI: 10.1200/po.23.00240] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/30/2023] [Accepted: 12/04/2023] [Indexed: 03/29/2025] Open
Abstract
PURPOSE Targeted Agent and Profiling Utilization Registry (TAPUR) is a phase II basket trial evaluating the antitumor activity of commercially available targeted agents in patients with advanced cancer and genomic alterations known to be drug targets. Results of a cohort of patients with advanced pancreatic cancer with BRCA1 /2 mutations treated with olaparib are reported. METHODS Eligible patients had advanced pancreatic cancer, measurable disease, Eastern Cooperative Oncology Group performance status 0-2, adequate organ function, and no standard treatment options available. Genomic testing was performed in Clinical Laboratory Improvement Amendments–certified, College of American Pathologists-accredited site selected laboratories. Simon's two-stage design was used with a primary end point of disease control (DC), defined as objective response (OR) or stable disease of at least 16 weeks duration (SD16+) according to RECIST v1.1. Secondary end points included OR, progression-free survival (PFS), overall survival (OS), duration of response, duration of stable disease, and safety. RESULTS Thirty patients with BRCA1 /2 mutations were enrolled from November 2016 to August 2019. The median number of reported previous therapies was 3 (range, 1-10). Two patients were not evaluable and excluded from efficacy analyses. Two patients with complete response, three with partial response and three with SD16+, were observed for DC and OR rates of 31% (90% CI, 18 to 40; P = .04) and 18% (95% CI, 6 to 37), respectively. The median PFS was 8 (95% CI, 8 to 15) weeks, and the median OS was 38 (95% CI, 21 to 65) weeks. Three patients had at least one drug-related grade 3 adverse event or serious adverse event of anemia, fever, or oral mucositis. CONCLUSION Olaparib showed antitumor activity in patients with advanced pancreatic cancer with BRCA1 /2 mutations extending findings of recent studies of olaparib in patients with this disease.
Collapse
Affiliation(s)
| | - Michael Rothe
- American Society of Clinical Oncology, Alexandria, VA
| | - Pam K. Mangat
- American Society of Clinical Oncology, Alexandria, VA
| | | | - Carmen J. Calfa
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | - Ajjai S. Alva
- University of Michigan Rogel Comprehensive Cancer Center, Ann Arbor, MI
| | - Vijay Suhag
- Sutter Health Roseville Cancer Center, Roseville, CA
| | | | | | - Omid Hamid
- The Angeles Clinic and Research Institute, A Cedars-Sinai Affiliate, Los Angeles, CA
| | - Eddy S. Yang
- Department of Radiation Oncology, O'Neal Comprehensive Cancer Center at the University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Cheng SM, Su YY, Chiang NJ, Wang CJ, Chao YJ, Huang CJ, Tsai HJ, Chen SH, Chang CY, Tsai CR, Li YJ, Yen CJ, Chuang SC, Chang JSM, Shan YS, Hwang DY, Chen LT. Germline mutations of homologous recombination genes and clinical outcomes in pancreatic cancer: a multicenter study in Taiwan. J Biomed Sci 2024; 31:21. [PMID: 38350919 PMCID: PMC10865564 DOI: 10.1186/s12929-024-01008-7] [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: 07/13/2023] [Accepted: 01/28/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Cancer susceptibility germline mutations are associated with pancreatic ductal adenocarcinoma (PDAC). However, the hereditary status of PDAC and its impact on survival is largely unknown in the Asian population. METHODS Exome sequencing was performed on 527 blood samples from PDAC individuals and analyzed for mutations in 80 oncogenic genes. Pathogenic and likely pathogenic (P/LP) germline variants were diagnosed according to the ACMG variant classification categories. The association between germline homologous recombination gene mutations (gHRmut, including BAP1, BRCA1, BRCA2, PALB2, ATM, BLM, BRIP1, CHEK2, NBN, MUTYH, FANCA and FANCC) and the treatment outcomes was explored in patients with stage III/IV diseases treated with first-line (1L) platinum-based versus platinum-free chemotherapy. RESULTS Overall, 104 of 527 (19.7%) patients carried germline P/LP variants. The most common mutated genes were BRCA2 (3.60%), followed by ATR (2.66%) and ATM (1.9%). After a median follow-up duration of 38.3-months (95% confidence interval, 95% CI 35.0-43.7), the median overall survival (OS) was not significantly different among patients with gHRmut, non-HR germline mutations, or no mutation (P = 0.43). Among the 320 patients with stage III/IV disease who received 1L combination chemotherapy, 32 (10%) had gHRmut. Of them, patients receiving 1L platinum-based chemotherapy exhibited a significantly longer median OS compared to those with platinum-free chemotherapy, 26.1 months (95% CI 12.7-33.7) versus 9.6 months (95% CI 5.9-17.6), P = 0.001. However, the median OS of patients without gHRmut was 14.5 months (95% CI 13.2-16.9) and 12.6 months (95% CI 10.8-14.7) for patients receiving 1L platinum-based and platinum-free chemotherapy, respectively (P = 0.22). These results were consistent after adjusting for potential confounding factors including age, tumor stage, performance status, and baseline CA 19.9 in the multivariate Cox regression analysis. CONCLUSIONS Our study showed that nearly 20% of Taiwanese PDAC patients carried germline P/LP variants. The longer survival observed in gHRmut patients treated with 1L platinum-based chemotherapy highlights the importance of germline testing for all patients with advanced PDAC at diagnosis.
Collapse
Affiliation(s)
- Siao Muk Cheng
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Yung-Yeh Su
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Deparment of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Internal Medicine, Kaohsiung Medical University Hospital and Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Nai-Jung Chiang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Jung Wang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Surgery, National Cheng-Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ying-Jui Chao
- Department of Surgery, National Cheng-Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Jui Huang
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Jen Tsai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Deparment of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Internal Medicine, Kaohsiung Medical University Hospital and Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shang-Hung Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Deparment of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Yen Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Chia-Rung Tsai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Yi-Jie Li
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Chia-Jui Yen
- Deparment of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Chang Chuang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jeffrey Shu-Ming Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Department of Surgery, National Cheng-Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Daw-Yang Hwang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Center for Biomarkers and Biotech Drugs, Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Precision Medicine Ph.D. Program, National Tsing Hua University, Hsinchu, Taiwan.
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.
- Deparment of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Department of Internal Medicine, Kaohsiung Medical University Hospital and Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
33
|
Tamaki A, Kato T, Sakurai Y, Sato K, Adachi K, Tadehara M, Kogami T, Matsushita M, Hoshino A, Sanoyama I, Numata Y, Umezawa A, Ichinoe M, Ichihara M, Kusano C, Murakumo Y. REV7 is involved in outcomes of platinum-based chemotherapy in pancreatic cancer by controlling the DNA damage response. Cancer Sci 2024; 115:660-671. [PMID: 38130032 PMCID: PMC10859597 DOI: 10.1111/cas.16044] [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: 09/05/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023] Open
Abstract
REV7 is a multifunctional protein implicated in various biological processes, including DNA damage response. REV7 expression in human cancer cells affects their sensitivity to DNA-damaging agents. In the present study, we investigated the significance of REV7 in pancreatic ductal adenocarcinoma (PDAC). REV7 expression was immunohistochemically examined in 92 resected PDAC specimens and 60 endoscopic ultrasound-guided fine-needle aspiration biopsy (EUS-FNAB) specimens of unresectable PDAC treated with platinum-based chemotherapy, and its association with clinicopathologic features was analyzed. Although REV7 expression was not significantly associated with the progression of primary tumors (T-factor and Stage) in either resected or unresectable PDAC, decreased levels of REV7 expression in EUS-FNAB specimens of unresectable PDAC were significantly associated with better outcomes of platinum-based chemotherapy and a favorable prognosis. REV7-deficient PDAC cell lines showed suppressed cell growth and enhanced sensitivity to cisplatin in vitro. Tumor-bearing mice generated using REV7-deficient PDAC cell lines also showed enhanced sensitivity to cisplatin in vivo. RNA sequencing analysis using WT and REV7-deficient PDAC cell lines revealed that REV7 inactivation promoted the downregulation of genes involved in the DNA repair and the upregulation of genes involved in apoptosis. Our results indicate that decreased expression of REV7 is associated with better outcomes of platinum-based chemotherapy in PDAC by suppressing the DNA damage response. It is also suggested that REV7 is a useful biomarker for predicting the outcome of platinum-based chemotherapy and the prognosis of unresectable PDAC and is a potential target for PDAC treatment.
Collapse
Affiliation(s)
- Akihiro Tamaki
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Takuya Kato
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yasutaka Sakurai
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Keita Sato
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Kai Adachi
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Masayoshi Tadehara
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Taro Kogami
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Masahiro Matsushita
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Akiyoshi Hoshino
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Itaru Sanoyama
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yoshiko Numata
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Atsuko Umezawa
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Masaaki Ichinoe
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Masatoshi Ichihara
- Department of Biomedical Sciences, College of Life and Health SciencesChubu UniversityKasugaiJapan
| | - Chika Kusano
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Yoshiki Murakumo
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| |
Collapse
|
34
|
Kubo T, Muramatsu J, Arihara Y, Murota A, Ishikawa K, Yoshida M, Nagashima H, Tamura F, Ikeda Y, Usami M, Ono M, Nakamura H, Watanabe D, Shibata T, Kasahara K, Sakurai A, Takada K. Clinical characterization of patients with gBRCA1/2 mutation-positive unresectable pancreatic cancer: a multicenter prospective study. Jpn J Clin Oncol 2024; 54:47-53. [PMID: 37791389 PMCID: PMC10773213 DOI: 10.1093/jjco/hyad131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Accumulating evidence has demonstrated platinum-based chemotherapy followed by maintenance therapy with a poly Adenosine diphosphate (ADP)-ribose polymerase inhibitor (olaparib) show benefits in unresectable pancreatic cancer with a germline (g)BRCA1/2 mutation. Evaluation of the germline BRCA1 and BRCA2 mutation is essential for making decisions on a treatment strategy for patients with unresectable pancreatic cancer. However, the detection rates of germline BRCA1 and BRCA2 mutations and efficacy of maintenance with olaparib remain undetermined, prospectively, in Japan. METHODS & RESULTS In this prospective analysis, the rate of germline BRCA1 and BRCA2 mutations and efficacy of chemotherapy were analyzed in 136 patients with pancreatic cancer who underwent BRACAnalysis® (85 patients) or FoundationOne® CDx (51 patients) between January 2020 and July 2022. A total of six patients (4.4%) had a germline BRCA1 and BRCA2 mutation. Five patients were treated with modified FOLFIRINOX and one with fluorouracil and oxaliplatin. All patients continued platinum-based chemotherapy for ˃4 months and were subsequently treated with olaparib as a maintenance therapy. The response rate to platinum-based chemotherapy in the germline BRCA1 and BRCA2 mutation-positive group was significantly better than that of the germline BRCA1 and BRCA2 mutation-negative group (66% vs 23%, P = 0.04). All patients harbouring a germline BRCA1 and BRCA2 mutation were able to switch to olaparib. The median progression-free survival using olaparib was 5.7 months (range 3.0-9.2). CONCLUSIONS The rate of germline BRCA1 and BRCA2 mutations found in patients with unresectable pancreatic cancer was comparable to those of previous studies.An analysis of germline BRCA1 and BRCA2 mutations has benefits for all patients with unresectable pancreatic cancer with regard to decisions on therapeutic strategies in a clinical practice setting.
Collapse
Affiliation(s)
- Tomohiro Kubo
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Joji Muramatsu
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yohei Arihara
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ayako Murota
- Department of Medical Genetics and Genomics, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kazuma Ishikawa
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Makoto Yoshida
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | - Fumito Tamura
- Department of Gastroenterology, Hokkaido Cancer Center, Sapporo, Japan
| | - Yuki Ikeda
- Department of Gastroenterology, Oji General Hospital, Tomakomai, Japan
| | - Makoto Usami
- Department of Medical Oncology, Steel Memorial Muroran Hospital, Muroran, Japan
| | - Michihiro Ono
- Department of Gastroenterology, Steel Memorial Muroran Hospital, Muroran, Japan
| | - Hajime Nakamura
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Gastroenterology, Otaru Ekisaikai Hospital, Otaru, Japan
| | - Daichi Watanabe
- Department of Gastroenterology, Japanese Red Cross Date Hospital, Date, Japan
| | - Takanori Shibata
- Department of Gastroenterology, Rumoi City Hospital, Rumoi, Japan
| | - Kaoru Kasahara
- Department of Gastroenterology, Hakodate Goryoukaku Hospital, Hakodate, Japan
| | - Akihiro Sakurai
- Department of Medical Genetics and Genomics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kohichi Takada
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
35
|
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a rising incidence and is one of the most lethal human malignancies. Much is known regarding the biology and pathophysiology of PDAC, but translating this knowledge to the clinic to improve patient outcomes has been challenging. In this Review, we discuss advances and practice-changing trials for PDAC. We briefly review therapeutic failures as well as ongoing research to refine the standard of care, including novel biomarkers and clinical trial designs. In addition, we highlight contemporary areas of research, including poly(ADP-ribose) polymerase inhibitors, KRAS-targeted therapies and immunotherapies. Finally, we discuss the future of pancreatic cancer research and areas for improvement in the next decade.
Collapse
Affiliation(s)
- Z Ian Hu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eileen M O'Reilly
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
36
|
Buckley CW, O’Reilly EM. Next-generation therapies for pancreatic cancer. Expert Rev Gastroenterol Hepatol 2024; 18:55-72. [PMID: 38415709 PMCID: PMC10960610 DOI: 10.1080/17474124.2024.2322648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024]
Abstract
INTRODUCTION Pancreas ductal adenocarcinoma (PDAC) is a frequently lethal malignancy that poses unique therapeutic challenges. The current mainstay of therapy for metastatic PDAC (mPDAC) is cytotoxic chemotherapy. NALIRIFOX (liposomal irinotecan, fluorouracil, leucovorin, oxaliplatin) is an emerging standard of care in the metastatic setting. An evolving understanding of PDAC pathogenesis is driving a shift toward targeted therapy. Olaparib, a poly-ADP-ribose polymerase (PARP) inhibitor, has regulatory approval for maintenance therapy in BRCA-mutated mPDAC along with other targeted agents receiving disease-agnostic approvals including for PDAC with rare fusions and mismatch repair deficiency. Ongoing research continues to identify and evaluate an expanding array of targeted therapies for PDAC. AREAS COVERED This review provides a brief overview of standard therapies for PDAC and an emphasis on current and emerging targeted therapies. EXPERT OPINION There is notable potential for targeted therapies for KRAS-mutated PDAC with opportunity for meaningful benefit for a sizable portion of patients with this disease. Further, emerging approaches are focused on novel immune, tumor microenvironment, and synthetic lethality strategies.
Collapse
Affiliation(s)
- Conor W. Buckley
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Eileen M. O’Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
- Weill Cornell Medicine, New York, USA
| |
Collapse
|
37
|
Pantaleo A, Forte G, Fasano C, Lepore Signorile M, Sanese P, De Marco K, Di Nicola E, Latrofa M, Grossi V, Disciglio V, Simone C. Understanding the Genetic Landscape of Pancreatic Ductal Adenocarcinoma to Support Personalized Medicine: A Systematic Review. Cancers (Basel) 2023; 16:56. [PMID: 38201484 PMCID: PMC10778202 DOI: 10.3390/cancers16010056] [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: 11/09/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal malignancies worldwide. While population-wide screening recommendations for PDAC in asymptomatic individuals are not achievable due to its relatively low incidence, pancreatic cancer surveillance programs are recommended for patients with germline causative variants in PDAC susceptibility genes or a strong family history. In this study, we sought to determine the prevalence and significance of germline alterations in major genes (ATM, BRCA1, BRCA2, CDKN2A, EPCAM, MLH1, MSH2, MSH6, PALB2, PMS2, STK11, TP53) involved in PDAC susceptibility. We performed a systematic review of PubMed publications reporting germline variants identified in these genes in PDAC patients. Overall, the retrieved articles included 1493 PDAC patients. A high proportion of these patients (n = 1225/1493, 82%) were found to harbor alterations in genes (ATM, BRCA1, BRCA2, PALB2) involved in the homologous recombination repair (HRR) pathway. Specifically, the remaining PDAC patients were reported to carry alterations in genes playing a role in other cancer pathways (CDKN2A, STK11, TP53; n = 181/1493, 12.1%) or in the mismatch repair (MMR) pathway (MLH1, MSH2, MSH6, PMS2; n = 87/1493, 5.8%). Our findings highlight the importance of germline genetic characterization in PDAC patients for better personalized targeted therapies, clinical management, and surveillance.
Collapse
Affiliation(s)
- Antonino Pantaleo
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Bari, Italy; (A.P.); (G.F.); (C.F.); (M.L.S.); (P.S.); (K.D.M.); (E.D.N.); (M.L.); (V.G.)
| | - Giovanna Forte
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Bari, Italy; (A.P.); (G.F.); (C.F.); (M.L.S.); (P.S.); (K.D.M.); (E.D.N.); (M.L.); (V.G.)
| | - Candida Fasano
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Bari, Italy; (A.P.); (G.F.); (C.F.); (M.L.S.); (P.S.); (K.D.M.); (E.D.N.); (M.L.); (V.G.)
| | - Martina Lepore Signorile
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Bari, Italy; (A.P.); (G.F.); (C.F.); (M.L.S.); (P.S.); (K.D.M.); (E.D.N.); (M.L.); (V.G.)
| | - Paola Sanese
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Bari, Italy; (A.P.); (G.F.); (C.F.); (M.L.S.); (P.S.); (K.D.M.); (E.D.N.); (M.L.); (V.G.)
| | - Katia De Marco
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Bari, Italy; (A.P.); (G.F.); (C.F.); (M.L.S.); (P.S.); (K.D.M.); (E.D.N.); (M.L.); (V.G.)
| | - Elisabetta Di Nicola
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Bari, Italy; (A.P.); (G.F.); (C.F.); (M.L.S.); (P.S.); (K.D.M.); (E.D.N.); (M.L.); (V.G.)
| | - Marialaura Latrofa
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Bari, Italy; (A.P.); (G.F.); (C.F.); (M.L.S.); (P.S.); (K.D.M.); (E.D.N.); (M.L.); (V.G.)
| | - Valentina Grossi
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Bari, Italy; (A.P.); (G.F.); (C.F.); (M.L.S.); (P.S.); (K.D.M.); (E.D.N.); (M.L.); (V.G.)
| | - Vittoria Disciglio
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Bari, Italy; (A.P.); (G.F.); (C.F.); (M.L.S.); (P.S.); (K.D.M.); (E.D.N.); (M.L.); (V.G.)
| | - Cristiano Simone
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Bari, Italy; (A.P.); (G.F.); (C.F.); (M.L.S.); (P.S.); (K.D.M.); (E.D.N.); (M.L.); (V.G.)
- Medical Genetics, Department of Precision and Regenerative Medicine and Jonic Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
38
|
Li XJ, Nie P, Herdewijn P, Sun JG. Unlocking the synthetic approaches and clinical application of approved small-molecule drugs for gastrointestinal cancer treatment: A comprehensive exploration. Eur J Med Chem 2023; 262:115928. [PMID: 37944387 DOI: 10.1016/j.ejmech.2023.115928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
Gastrointestinal (GI) cancers encompass a group of malignancies affecting the digestive system, including the stomach, esophagus, liver, colon, rectum and pancreas. These cancers represent a significant global health burden, necessitating effective treatment strategies. Small-molecule drugs have emerged as crucial therapeutic options in the fight against GI cancers due to their oral bioavailability, targeted mechanisms of action, and well-established safety profiles. The review then elucidates the clinical applications and synthetic methods of clinically approved small-molecule drugs for the treatment of GI cancer, shedding light on their mechanisms of action and their potential in mitigating GI cancer progression. The review also discusses future prospects and the evolving landscape of small-molecule drug development in GI oncology, highlighting the potential for personalized medicine. In summary, this review provides valuable insights into cutting-edge strategies for harnessing clinically approved small-molecule drugs to combat GI cancer effectively.
Collapse
Affiliation(s)
- Xiao-Jing Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Peng Nie
- Medicinal Chemistry, Rega Institute of Medical Research, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Piet Herdewijn
- Medicinal Chemistry, Rega Institute of Medical Research, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Jian-Gang Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
39
|
Kitamura H, Morizane C, Tanabe N, Go I, Maruki Y, Ohba A, Nagashio Y, Kondo S, Hijioka S, Ueno H, Yoshida T, Okusaka T. Clinical features of germline BRCA1/2 or ATM pathogenic variant positive pancreatic cancer in Japan. Pancreatology 2023; 23:964-969. [PMID: 37914629 DOI: 10.1016/j.pan.2023.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/08/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND There has been increasing interest into the role of germline BRCA1/2 pathogenic variants (gBRCA PV) and gATM PV and likely PV (PV and LPV; PV + LPV) in the carcinogenesis and treatment of pancreatic cancer (PC), but the clinical features have not been well described. METHODS Patients with confirmed gBRCA PV and gATM PV + LPV PC treated at our hospital between April 2016 and December 2021, were retrospectively evaluated for clinical characteristics and outcomes. RESULTS Twenty-two patients harbored gBRCA PV and three patients harbored gATM PV + LPV. Of the gBRCA PV patients, 81.8 % received platinum-based chemotherapy with favorable treatment outcomes with an objective response rate of 50.0 % (95 % CI: 23.0-77.0), median progression free survival (PFS) of 334 days, and median overall survival (OS) of 926 days from the initiation of first-line chemotherapy. The annual number of patients with gBRCA PV was two patients per year before January 2021 (when BRACAnalysis became available in Japan), and ten patients during the 10 months thereafter. Four patients (20 %) with gBRCA PV developed soft-tissue metastasis with progression. Two patients with gATM PV + LPV received platinum-based chemotherapy and the best response of those patients was partial response and stable disease and their OS from the initiation of first-line chemotherapy was 1192 and 989 days, and PFS was 579 and 140 days, respectively. CONCLUSION The diagnosis of gBRCA PV-positive PC has increased revealed in recent years. These tumors appear to be sensitive to platinum-based chemotherapy, with long term survival observed in gATM PV + LPV-positive patients.
Collapse
Affiliation(s)
- Hidetoshi Kitamura
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan.
| | - Noriko Tanabe
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Japan
| | - Ikeda Go
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Yuta Maruki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Akihiro Ohba
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Yoshikuni Nagashio
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Shunsuke Kondo
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Susumu Hijioka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Hideki Ueno
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Teruhiko Yoshida
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| |
Collapse
|
40
|
Xing J, Ge Y, Gong X, Liu Y, Cheng Y. Initial chemotherapy option for pancreatic ductal adenocarcinoma in patients with adequate performance status. JOURNAL OF PANCREATOLOGY 2023; 6:196-201. [DOI: 10.1097/jp9.0000000000000144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly progressive lethal malignancy, with chemotherapy being the primary treatment modality. This article provides a review of the initial chemotherapy options for PDAC patients with adequate performance status, comparing FOLFIRINOX (oxaliplatin, irinotecan, 5-fluorouracil, and leucovorin) or modified FOLFIRINOX and gemcitabine plus nab-paclitaxel (GEM-NabP) regimens. The availability of limited evidence from randomized trials restricts a direct comparison between the 2 regimens. Based on our review, (m)FOLFIRINOX yields superior survival outcomes compared to GEM-NabP in metastatic PDAC. For locally advanced PDAC, either (m)FOLFIRINOX or GEM-NabP can be considered initial chemotherapy. In the neoadjuvant setting for borderline resectable PDAC, both regimens have demonstrated promising results in achieving feasible resection rates. However, mFOLFIRINOX remains the preferred choice for adjuvant chemotherapy. The selection of initial chemotherapy for PDAC depends on the disease stage, patients’ performance status, and tumor molecular alterations. Further research and clinical trials are necessary to optimize treatment approaches for PDAC patients.
Collapse
Affiliation(s)
- Jiazhang Xing
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuping Ge
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaolei Gong
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Liu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
41
|
Militello AM, Orsi G, Cavaliere A, Niger M, Avallone A, Salvatore L, Tortora G, Rapposelli IG, Giordano G, Noventa S, Giommoni E, Bozzarelli S, Macchini M, Peretti U, Procaccio L, Puccini A, Cascinu S, Montagna C, Milella M, Reni M. Clinical outcomes and response to chemotherapy in a cohort of pancreatic cancer patients with germline variants of unknown significance (VUS) in BRCA1 and BRCA2 genes. Cancer Chemother Pharmacol 2023; 92:501-510. [PMID: 37725113 DOI: 10.1007/s00280-023-04585-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 08/22/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE The clinical outcome and the efficacy of chemotherapy in pancreatic cancer patients with BRCA1/2 Variants of Unknown Significance (VUS) is unknown. We explored the effects of chemotherapy with or without Platinum in non metastatic and metastatic pancreatic cancer patients with BRCA1/2 VUS. METHODS A retrospective analysis of non-metastatic or metastatic pancreatic cancer patients with gBRCA1/2 VUS treated in 13 Italian centers between November 2015 and December 2020 was performed. All patients were assessed for toxicity and RECIST 1.1 response. Metastatic patients were evaluated for survival outcome. RESULTS 30 pancreatic cancer patients with gBRCA1/2 VUS were considered: 20 were M+ and 10 were non-M+. Pl-CT was recommended to 16 patients: 10 M+ (6 FOLFIRINOX and 4 PAXG) and 6 non-M+ (3 FOLFIRINOX and 3 PAXG); 11 patients received Nabpaclitaxel-Gemcitabine (AG; 8 M+) and 3 patients (2 M+) were treated with Gemcitabine (G). The RECIST 1.1 response rate was 27% for AG and 44% for Pl-CT (22% for (m) FOLFIRINOX and 71% PAXG). 1 year Progression-Free Survival was 37.5% for patients treated with AG and 33% in the Pl-CT subgroup. Median Overall Survival (OS) was 23.5 months for patients treated with AG and 14 months for the Pl-CT subgroup. 1 Year and 2 Year OS were numerically better for AG (1 Year OS: 75% vs 60% and 2 Year OS: 50% and 20% in AG and Pl-CT subgroups, respectively) as well. CONCLUSIONS Pl-CT does not seem to be associated with a better outcome compared to AG chemotherapy in PDAC patients with BRCA 1/2 VUS.
Collapse
Affiliation(s)
- Anna Maria Militello
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Orsi
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Cavaliere
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO - IRCCS Candiolo, Candiolo, Italy
| | - Monica Niger
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Antonio Avallone
- Biologia Cellulare e Bioterapie, Istituto Nazionale per lo Studio e la Cura dei Tumori ''Fondazione Giovanni Pascale'' - IRCCS, Naples, Italy
| | - Lisa Salvatore
- Unit of Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario, Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giampaolo Tortora
- Unit of Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario, Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ilario Giovanni Rapposelli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ''Dino Amadori'', Meldola, Italy
| | - Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy, Policlinico Riuniti, Foggia, Italy
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Silvia Noventa
- Department of Medical Oncology, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Elisa Giommoni
- Medical Oncology Division, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Silvia Bozzarelli
- Department of Medical Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Marina Macchini
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Umberto Peretti
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Letizia Procaccio
- Medical Oncology 1 Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Alberto Puccini
- University of Genoa, Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Stefano Cascinu
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Montagna
- Department of Radiation Oncology and Genomic Instability and Cancer Genetics, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Michele Reni
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 60, 20123, Milan, Italy.
- Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
42
|
Lim DH, Yoon H, Kim KP, Ryoo BY, Lee SS, Park DH, Song TJ, Hwang DW, Lee JH, Song KB, Kim SC, Hong SM, Hyung J, Yoo C. Analysis of Plasma Circulating Tumor DNA in Borderline Resectable Pancreatic Cancer Treated with Neoadjuvant Modified FOLFIRINOX: Clinical Relevance of DNA Damage Repair Gene Alteration Detection. Cancer Res Treat 2023; 55:1313-1320. [PMID: 37139665 PMCID: PMC10582539 DOI: 10.4143/crt.2023.452] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/04/2023] [Indexed: 05/05/2023] Open
Abstract
PURPOSE There are no reliable biomarkers to guide treatment for patients with borderline resectable pancreatic cancer (BRPC) in the neoadjuvant setting. We used plasma circulating tumor DNA (ctDNA) sequencing to search biomarkers for patients with BRPC receiving neoadjuvant mFOLFIRINOX in our phase 2 clinical trial (NCT02749136). MATERIALS AND METHODS Among the 44 patients enrolled in the trial, patients with plasma ctDNA sequencing at baseline or post-operation were included in this analysis. Plasma cell-free DNA isolation and sequencing were performed using the Guardant 360 assay. Detection of genomic alterations, including DNA damage repair (DDR) genes, were examined for correlations with survival. RESULTS Among the 44 patients, 28 patients had ctDNA sequencing data qualified for the analysis and were included in this study. Among the 25 patients with baseline plasma ctDNA data, 10 patients (40%) had alterations of DDR genes detected at baseline, inclu-ding ATM, BRCA1, BRCA2 and MLH1, and showed significantly better progression-free survival than those without such DDR gene alterations detected (median, 26.6 vs. 13.5 months; log-rank p=0.004). Patients with somatic KRAS mutations detected at baseline (n=6) had significantly worse overall survival (median, 8.5 months vs. not applicable; log-rank p=0.003) than those without. Among 13 patients with post-operative plasma ctDNA data, eight patients (61.5%) had detectable somatic alterations. CONCLUSION Detection of DDR gene mutations from plasma ctDNA at baseline was associated with better survival outcomes of pati-ents with borderline resectable pancreatic ductal adenocarcinoma treated with neoadjuvant mFOLFIRINOX and may be a prognostic biomarker.
Collapse
Affiliation(s)
- Dong-Hoon Lim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Hyunseok Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Kyu-pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Sang Soo Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Do Hyun Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Tae Jun Song
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Dae Wook Hwang
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Jae Hoon Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Ki Byung Song
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Song Cheol Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Jaewon Hyung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| |
Collapse
|
43
|
Lee E, Archasappawat S, Ji K, Pena J, Fernandez-Vega V, Gangaraju R, Beesabathuni NS, Kim MJ, Tian Q, Shah PS, Scampavia L, Spicer TP, Hwang CI. A new vulnerability to BET inhibition due to enhanced autophagy in BRCA2 deficient pancreatic cancer. Cell Death Dis 2023; 14:620. [PMID: 37735513 PMCID: PMC10514057 DOI: 10.1038/s41419-023-06145-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023]
Abstract
Pancreatic cancer is one of the deadliest diseases in human malignancies. Among total pancreatic cancer patients, ~10% of patients are categorized as familial pancreatic cancer (FPC) patients, carrying germline mutations of the genes involved in DNA repair pathways (e.g., BRCA2). Personalized medicine approaches tailored toward patients' mutations would improve patients' outcome. To identify novel vulnerabilities of BRCA2-deficient pancreatic cancer, we generated isogenic Brca2-deficient murine pancreatic cancer cell lines and performed high-throughput drug screens. High-throughput drug screening revealed that Brca2-deficient cells are sensitive to Bromodomain and Extraterminal Motif (BET) inhibitors, suggesting that BET inhibition might be a potential therapeutic approach. We found that BRCA2 deficiency increased autophagic flux, which was further enhanced by BET inhibition in Brca2-deficient pancreatic cancer cells, resulting in autophagy-dependent cell death. Our data suggests that BET inhibition can be a novel therapeutic strategy for BRCA2-deficient pancreatic cancer.
Collapse
Affiliation(s)
- EunJung Lee
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Suyakarn Archasappawat
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
- Graduate Group in Integrative Pathobiology, University of California, Davis, Davis, CA, 95616, USA
| | - Keely Ji
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Jocelyn Pena
- The Herbert Wertheim UF Scripps Institute, High-Throughput Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL, 33458, USA
| | - Virneliz Fernandez-Vega
- The Herbert Wertheim UF Scripps Institute, High-Throughput Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL, 33458, USA
| | - Ritika Gangaraju
- Department of Chemical Engineering, College of Engineering, University of California, Davis, Davis, CA, 95616, USA
| | - Nitin Sai Beesabathuni
- Department of Chemical Engineering, College of Engineering, University of California, Davis, Davis, CA, 95616, USA
| | - Martin Jean Kim
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Qi Tian
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Priya S Shah
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
- Department of Chemical Engineering, College of Engineering, University of California, Davis, Davis, CA, 95616, USA
| | - Louis Scampavia
- The Herbert Wertheim UF Scripps Institute, High-Throughput Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL, 33458, USA
| | - Timothy P Spicer
- The Herbert Wertheim UF Scripps Institute, High-Throughput Screening Center, Department of Molecular Medicine, Scripps Florida, Jupiter, FL, 33458, USA
| | - Chang-Il Hwang
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA.
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
44
|
Golan T, Casolino R, Biankin AV, Hammel P, Whitaker KD, Hall MJ, Riegert-Johnson DL. Germline BRCA testing in pancreatic cancer: improving awareness, timing, turnaround, and uptake. Ther Adv Med Oncol 2023; 15:17588359231189127. [PMID: 37720496 PMCID: PMC10504836 DOI: 10.1177/17588359231189127] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 07/04/2023] [Indexed: 09/19/2023] Open
Abstract
Prognosis is generally poor for patients with pancreatic ductal adenocarcinoma. However, patients with germline BRCA1 or BRCA2 mutations (gBRCAm) may benefit from first-line platinum-based chemotherapy and maintenance therapy with the poly(adenosine diphosphate-ribose) polymerase inhibitor olaparib following at least 16 weeks of first-line platinum-based chemotherapy without disease progression. Germline breast cancer gene (BRCA) testing is therefore important to ensure that patients receive the most effective treatment. In addition, testing for other DNA damage response gene mutations beyond gBRCAm may also guide treatment decisions. However, clinical pathways for genetic testing are often suboptimal, leading to delays in treatment initiation or missed opportunities for personalized therapy. Barriers to testing include low rates of referral and uptake, delays to referral and slow result turnaround times, cost, and biopsy and assay limitations if somatic testing is performed, leading to the requirement for subsequent dedicated germline testing. Low rates of referral may result from lack of awareness among physicians of the clinical value of testing, coupled with low confidence in interpreting test results and poor availability of genetic counseling services. Among patients, barriers to uptake may include similar lack of awareness of the clinical value of testing, anxiety regarding the implications of test results, lack of insurance coverage, fear of negative insurance implications, and socioeconomic factors. Potential solutions include innovative approaches to testing pathways, including 'mainstreaming' of testing in which BRCA tests are routinely arranged by the treating oncologist, with the involvement of genetic counselors if a patient is found to have a gBRCAm. More recently, the utility of multigene panel analyses has also been explored. Access to genetic counseling may also be improved through initiatives such as having a genetic counseling appointment for all new patient visits and telemedicine approaches, including the use of telephone consultations or DVD-assisted counseling. Educational programs will also be beneficial, and cost effectiveness is likely to improve as the number of targeted treatments increases and when the earlier detection of tumors in family members following cascade testing is considered.
Collapse
Affiliation(s)
- Talia Golan
- Institute of Oncology, Sheba Medical Center, Tel Hashomer 52621, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Raffaella Casolino
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Andrew V. Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Liverpool, Australia
| | - Pascal Hammel
- Department of Digestive and Medical Oncology, University Paris-Saclay, Paul Brousse Hospital (AP-HP), Villejuif, France
| | - Kristen D. Whitaker
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Michael J. Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | |
Collapse
|
45
|
Asiri MS, Dabaliz A, Almutairi M, Almahbub A, Alharbi M, Almeman S, AlShieban S, Alotaibi T, Algarni M. Complete Pathological Response to Platinum-Based Neoadjuvant Chemotherapy in BRCA2-Associated Locally Advanced Pancreatic Cancer: A Case Report and Literature Review. Cureus 2023; 15:e43261. [PMID: 37692681 PMCID: PMC10492221 DOI: 10.7759/cureus.43261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignant disease and is considered the fourth leading cause of death among cancer patients in the United States. Mutations in the BRCA gene, which is a DNA repair gene, increase the risk of PDAC, and among all patients with PDAC, about 8%-10% have a BRCA2 mutation. The finding of gene mutations is associated with a better response to platinum-based chemotherapy. Here, we present a case of a 59-year-old male with a BRCA2 gene mutation who was diagnosed with locally advanced pancreatic cancer and had achieved a complete pathological response to the FOLFIRINOX (leucovorin calcium, fluorouracil, irinotecan hydrochloride, and oxaliplatin) regimen and Whipple procedure. We also present our literature findings on response types in BRCA2 PDAC patients, as well as consensus on the use of different therapies. The use of platinum-based chemotherapy with BRCA2 is highly recommended as the first-line treatment. Most PDAC patients remain untested for BRCA2 mutation even though their genetic status influences the selection of drug regimens. Thus, we recommend genetic testing for everyone with PDAC.
Collapse
Affiliation(s)
- Mohamed S Asiri
- Medicine, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
- Medicine, King Abdullah International Medical Research Center, Riyadh, SAU
| | - Alhomam Dabaliz
- Medicine, College of Medicine, Alfaisal University, Riyadh, SAU
| | - Mahdi Almutairi
- Medicine, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
| | - Abdulaziz Almahbub
- Medicine, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
| | - Mohammed Alharbi
- Pathology and Laboratory Medicine, King Abdulaziz Medical City, Riyadh, Riyadh, SAU
| | - Sarah Almeman
- Pathology and Laboratory Medicine, King Abdulaziz Medical City, Riyadh, Riyadh, SAU
| | - Saeed AlShieban
- Pathology and Laboratory Medicine, King Abdulaziz Medical City, Riyadh, Riyadh, SAU
- Pathology and Laboratory Medicine, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
- Pathology and Laboratory Medicine, King Abdullah International Medical Research Center, Riyadh, SAU
| | - Tareq Alotaibi
- Medical Imaging, King Abdulaziz Medical City, Riyadh, Riyadh, SAU
| | - Mohammed Algarni
- Oncology, King Abdulaziz Medical City, Riyadh, Riyadh, SAU
- Oncology, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
- Oncology, King Abdullah International Medical Research Center, Riyadh, SAU
| |
Collapse
|
46
|
Lee PWP, Strum SW, Tsvetkova E. It Is All in the Genes: A Story of Unexpected Survival in a 67-Year-Old Male with Metastatic Pancreatic Cancer. Case Rep Oncol Med 2023; 2023:8751205. [PMID: 37547629 PMCID: PMC10400294 DOI: 10.1155/2023/8751205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/07/2023] [Indexed: 08/08/2023] Open
Abstract
Background We describe a case report of a 67-year-old male with PDAC who experienced an exceptional survival outcome during systemic therapy and its implications in precision medicine. We hypothesize that his outcomes are attributable, in part, to a germline BRCA2 deletion and somatic GNAS substitution. Methods Retrospective single-patient chart review was performed at the London Regional Cancer Program, as well as a structured literature search spanning all years in PubMed of BRCA and GNAS mutations in pancreatic cancer. Results The case described herein represents a 67-year-old male who survived over 27 months after third-line treatment with gemcitabine, docetaxel, and capecitabine (GTX) chemotherapy for metastatic PDAC after progression on gemcitabine and Abraxane and then on FOLFIRINOX. His survival far exceeded the median overall survival metrics. Genetic testing revealed a pathogenic heterozygous germline BRCA2 6643delT p.(Tyr2215Thrfs∗14) frameshift mutation and somatic GNAS 2531G > A p.(Arg844His) mutation. Conclusions This case highlights the urgent need to expand our knowledge of cancer biology to advance personalized cancer treatment and therapy development.
Collapse
Affiliation(s)
- Patsy W. P. Lee
- Department of Internal Medicine, Schulich School of Medicine and Dentistry, Western University, Canada
| | - Scott W. Strum
- Department of Medical Oncology, London Regional Cancer Program, Western University, Canada
| | - Elena Tsvetkova
- Department of Medical Oncology, London Regional Cancer Program, Western University, Canada
| |
Collapse
|
47
|
Abstract
Biomolecular condensates are reversible compartments that form through a process called phase separation. Post-translational modifications like ADP-ribosylation can nucleate the formation of these condensates by accelerating the self-association of proteins. Poly(ADP-ribose) (PAR) chains are remarkably transient modifications with turnover rates on the order of minutes, yet they can be required for the formation of granules in response to oxidative stress, DNA damage, and other stimuli. Moreover, accumulation of PAR is linked with adverse phase transitions in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. In this review, we provide a primer on how PAR is synthesized and regulated, the diverse structures and chemistries of ADP-ribosylation modifications, and protein-PAR interactions. We review substantial progress in recent efforts to determine the molecular mechanism of PAR-mediated phase separation, and we further delineate how inhibitors of PAR polymerases may be effective treatments for neurodegenerative pathologies. Finally, we highlight the need for rigorous biochemical interrogation of ADP-ribosylation in vivo and in vitro to clarify the exact pathway from PARylation to condensate formation.
Collapse
Affiliation(s)
- Kevin Rhine
- Program in Cell, Molecular, Developmental Biology, and Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Hana M Odeh
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - James Shorter
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Sua Myong
- Program in Cell, Molecular, Developmental Biology, and Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Physics Frontier Center (Center for the Physics of Living Cells), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
48
|
Calheiros J, Corbo V, Saraiva L. Overcoming therapeutic resistance in pancreatic cancer: Emerging opportunities by targeting BRCAs and p53. Biochim Biophys Acta Rev Cancer 2023; 1878:188914. [PMID: 37201730 DOI: 10.1016/j.bbcan.2023.188914] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023]
Abstract
Pancreatic cancer (PC) is characterized by (epi)genetic and microenvironmental alterations that negatively impact the treatment outcomes. New targeted therapies have been pursued to counteract the therapeutic resistance in PC. Aiming to seek for new therapeutic options for PC, several attempts have been undertaken to exploit BRCA1/2 and TP53 deficiencies as promising actionable targets. The elucidation of the pathogenesis of PC highlighted the high prevalence of p53 mutations and their connection with the aggressiveness and therapeutic resistance of PC. Additionally, PC is associated with dysfunctions in several DNA repair-related genes, including BRCA1/2, which sensitize tumours to DNA-damaging agents. In this context, poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) were approved for mutant BRCA1/2 PC patients. However, acquired drug resistance has become a major drawback of PARPi. This review emphasizes the importance of targeting defective BRCAs and p53 pathways for advancing personalized PC therapy, with particular focus on how this approach may provide an opportunity to tackle PC resistance.
Collapse
Affiliation(s)
- Juliana Calheiros
- LAQV/REQUIMTE, Laboratόrio de Microbiologia, Departamento de Ciências Biolόgicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine (DIMI), University and Hospital Trust of Verona, Verona, Italy; ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratόrio de Microbiologia, Departamento de Ciências Biolόgicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
49
|
Schepis T, De Lucia SS, Pellegrino A, del Gaudio A, Maresca R, Coppola G, Chiappetta MF, Gasbarrini A, Franceschi F, Candelli M, Nista EC. State-of-the-Art and Upcoming Innovations in Pancreatic Cancer Care: A Step Forward to Precision Medicine. Cancers (Basel) 2023; 15:3423. [PMID: 37444534 PMCID: PMC10341055 DOI: 10.3390/cancers15133423] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Pancreatic cancer remains a social and medical burden despite the tremendous advances that medicine has made in the last two decades. The incidence of pancreatic cancer is increasing, and it continues to be associated with high mortality and morbidity rates. The difficulty of early diagnosis (the lack of specific symptoms and biomarkers at early stages), the aggressiveness of the disease, and its resistance to systemic therapies are the main factors for the poor prognosis of pancreatic cancer. The only curative treatment for pancreatic cancer is surgery, but the vast majority of patients with pancreatic cancer have advanced disease at the time of diagnosis. Pancreatic surgery is among the most challenging surgical procedures, but recent improvements in surgical techniques, careful patient selection, and the availability of minimally invasive techniques (e.g., robotic surgery) have dramatically reduced the morbidity and mortality associated with pancreatic surgery. Patients who are not candidates for surgery may benefit from locoregional and systemic therapy. In some cases (e.g., patients for whom marginal resection is feasible), systemic therapy may be considered a bridge to surgery to allow downstaging of the cancer; in other cases (e.g., metastatic disease), systemic therapy is considered the standard approach with the goal of prolonging patient survival. The complexity of patients with pancreatic cancer requires a personalized and multidisciplinary approach to choose the best treatment for each clinical situation. The aim of this article is to provide a literature review of the available treatments for the different stages of pancreatic cancer.
Collapse
Affiliation(s)
- Tommaso Schepis
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Sara Sofia De Lucia
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Antonio Pellegrino
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Angelo del Gaudio
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Rossella Maresca
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Gaetano Coppola
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Michele Francesco Chiappetta
- Section of Gastroenterology and Hepatology, Promise, Policlinico Universitario Paolo Giaccone, 90127 Palermo, Italy;
- IBD-Unit, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Antonio Gasbarrini
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Anesthesiological and Reanimation Sciences, Fondazione Universitaria Policlinico Agostino Gemelli di Roma, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy; (F.F.); (M.C.)
| | - Marcello Candelli
- Department of Emergency Anesthesiological and Reanimation Sciences, Fondazione Universitaria Policlinico Agostino Gemelli di Roma, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy; (F.F.); (M.C.)
| | - Enrico Celestino Nista
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| |
Collapse
|
50
|
Vannini I, Rossi T, Melloni M, Valgiusti M, Urbini M, Passardi A, Bartolini G, Gallio C, Azzali I, Bandini S, Ancarani V, Montanaro L, Frassineti GL, Fabbri F, Rapposelli IG. Analysis of EVs from patients with advanced pancreatic cancer identifies antigens and miRNAs with predictive value. Mol Ther Methods Clin Dev 2023; 29:473-482. [PMID: 37273899 PMCID: PMC10238807 DOI: 10.1016/j.omtm.2023.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 05/09/2023] [Indexed: 06/06/2023]
Abstract
The identification of predictive factors for treatment of pancreatic cancer (PC) is an unmet clinical need. In the present work, we analyzed blood-derived extracellular vesicles (EVs) from patients with advanced PC in order to find a molecular signature predictive of response to therapy. We analyzed samples from 21 patients with advanced PC, all receiving first-line treatment with gemcitabine + nab-paclitaxel. Isolated EVs have been analyzed, and the results of laboratory have been matched with clinical data in order to investigate possible predictive factors. EV concentration and size were similar between responder and non-responder patients. Analysis of 37 EV surface epitopes showed a decreased expression of SSEA4 and CD81 in responder patients. We detected more than 450 expressed miRNAs in EVs. A comparative survey between responder and non-responder patients showed that at least 44 miRNAs were differently expressed. Some of these miRNAs have already been observed in relation to the survival and gemcitabine sensitivity of tumor cells. In conclusion, we showed the ability of our approach to identify EV-derived biomarkers with predictive value for therapy response in PC. Our findings are worthy of further investigation, including the analysis of samples from patients treated with different schedules and in different settings.
Collapse
Affiliation(s)
- Ivan Vannini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Tania Rossi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Mattia Melloni
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Martina Valgiusti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Milena Urbini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giulia Bartolini
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Gallio
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Irene Azzali
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Valentina Ancarani
- Immunotherapy-Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Lorenzo Montanaro
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, Bologna, Italy
- Departmental Program in Laboratory Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Francesco Fabbri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ilario Giovanni Rapposelli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|