1
|
Sun X, Song H, Sun X, Liao C, Wang G, Xu Y, Li L, Han Y, Xu C, Wang W, Cai S, Liang H, Yu H. A 15-Inflammation-Related Gene Signature Predicts the Prognosis of Patients With Pancreatic Ductal Adenocarcinoma. Cancer Invest 2024:1-17. [PMID: 38616304 DOI: 10.1080/07357907.2024.2340577] [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: 11/14/2023] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
Chronic inflammation promotes the development of pancreatic ductal adenocarcinoma (PDAC) and PDAC-related inflammatory tumor microenvironment facilitates tumor growth and metastasis. Thus, we aimed to study the association between inflammatory response and prognosis in patients with PDAC. We conducted the whole transcriptomic sequencing using tissue samples collected from patients diagnosed with PDAC (n = 106) recruited from Shandong Cancer Hospital. We first constructed a prognostic signature using 15 inflammation-related genes in The Cancer Genome Atlas (TCGA) cohort (n = 177) and further validated it in an independent International Cancer Genome Consortium (ICGC) cohort (n = 90) and our in-house cohort. PDAC patients with a higher risk score had poorer overall survival (OS) (P < 0.001; HR, 3.02; 95% CI, 1.94-4.70). The association between the prognostic signature and OS remained significant in the multivariable Cox regression adjusting for age, sex, alcohol exposure, diabetes, and stage (P < 0.001; HR, 2.91; 95% CI, 1.73-4.89). This gene signature also robustly predicted prognosis in the ICGC cohort (P = 0.01; HR, 1.94; 95% CI, 1.14-3.30) and our cohort (P < 0.001; HR, 2.40; 95% CI, 1.45-3.97). Immune subtype C3 (inflammatory) was enriched and CD8+ T cells were higher in patients with a lower risk score (P < 0.05). Furthermore, PDAC patients with higher risk scores were more sensitive to chemotherapy, immunotherapy, and PARP inhibitors (P < 0.05). In sum, we identified a novel gene signature that was associated with inflammatory response for risk stratification, prognosis prediction, and therapy guidance in PDAC patients. Future studies are warranted to validate the clinical utility of the signature.
Collapse
Affiliation(s)
| | - Hao Song
- The Third Affiliated Hospital of Naval Military Medical University, Shanghai, P. R. China
| | - Xiaoran Sun
- Burning Rock Biotech, Guangzhou, P. R. China
| | | | | | - Yu Xu
- Burning Rock Biotech, Guangzhou, P. R. China
| | - Leo Li
- Burning Rock Biotech, Guangzhou, P. R. China
| | - Yusheng Han
- Burning Rock Biotech, Guangzhou, P. R. China
| | - Chunwei Xu
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, P. R. China
| | - Wenxian Wang
- The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, P. R. China
| | - Shangli Cai
- Burning Rock Biotech, Guangzhou, P. R. China
| | - Hua Liang
- Qingdao Central Hospital, Qingdao, P. R. China
| | - Hao Yu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, P. R. China
| |
Collapse
|
2
|
De La Fouchardière C, Malka D, Cropet C, Chabaud S, Raimbourg J, Botsen D, Launay S, Evesque L, Vienot A, Perrier H, Jary M, Rinaldi Y, Coutzac C, Bachet JB, Neuzillet C, Williet N, Desgrippes R, Grainville T, Aparicio T, Peytier A, Lecomte T, Roth GS, Thirot-Bidault A, Lachaux N, Bouché O, Ghiringhelli F. Gemcitabine and Paclitaxel Versus Gemcitabine Alone After 5-Fluorouracil, Oxaliplatin, and Irinotecan in Metastatic Pancreatic Adenocarcinoma: A Randomized Phase III PRODIGE 65-UCGI 36-GEMPAX UNICANCER Study. J Clin Oncol 2024; 42:1055-1066. [PMID: 38232341 DOI: 10.1200/jco.23.00795] [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: 04/11/2023] [Revised: 08/09/2023] [Accepted: 10/04/2023] [Indexed: 01/19/2024] Open
Abstract
PURPOSE GEMPAX was an open-label, randomized phase III clinical trial designed to assess the efficacy and tolerability of gemcitabine plus paclitaxel versus gemcitabine alone as second-line treatment for patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) who previously received 5-fluorouracil, oxaliplatin, and irinotecan. METHODS Patients with histologically or cytologically confirmed mPDAC were randomly assigned (2:1) to receive GEMPAX (paclitaxel 80 mg/m2 + gemcitabine 1,000 mg/m2; IV; once at day (D) 1, D8, and D15/arm A) or gemcitabine (arm B) alone once at D1, D8, and D15 every 28 days until progression, toxicity, or patient's decision. The primary end point was overall survival (OS). Secondary end points included progression-free survival (PFS), objective response rate (ORR), quality of life, and safety. RESULTS Overall, 211 patients (median age, 64 [30-86] years; 62% male) were included. After a median study follow-up for alive patients of 13.4 versus 13.8 months in arm A versus arm B, the median OS (95% CI) was 6.4 (5.2 to 7.4) versus 5.9 months (4.6 to 6.9; hazard ratio [HR], 0.87 [0.63 to 1.20]; P = 0.4095), the median PFS was 3.1 (2.2 to 4.3) versus 2.0 months (1.9 to 2.3; HR, 0.64 [0.47 to 0.89]; P = 0.0067), and the ORR was 17.1% (11.3 to 24.4) versus 4.2% (0.9 to 11.9; P = 0.008) in arm A versus arm B, respectively. Overall, 16.7% of patients in arm A and 2.9% in arm B discontinued their treatment because of adverse events (AEs). One grade 5 AE associated with both gemcitabine and paclitaxel was reported in arm A (acute respiratory distress), and 58.0% versus 27.1% of patients experienced grade ≥3 treatment-related AEs in arm A versus arm B, among which 15.2% versus 4.3% had anemia, 15.9% versus 15.7% had neutropenia, 19.6% versus 4.3% had thrombocytopenia, 10.1% versus 2.9% had asthenia and 12.3% versus 0.0% had neuropathy. CONCLUSION While GEMPAX did not meet the primary end point of OS versus gemcitabine alone in patients with mPDAC in the second-line setting, both PFS and ORR were significantly improved.
Collapse
Affiliation(s)
| | - David Malka
- Department of Cancer Medicine, Gustave Roussy, University of Paris-Saclay, Villejuif, France
| | - Claire Cropet
- Department of Clinical Research and Innovation, Léon Bérard Centre, Lyon, France
| | - Sylvie Chabaud
- Department of Clinical Research and Innovation, Léon Bérard Centre, Lyon, France
| | - Judith Raimbourg
- Medical Oncology Department, ICO René Gauducheau, Nantes, France
| | - Damien Botsen
- Medical Oncology Department, Godinot Institute, Unicancer Champagne, Reims, France
| | - Simon Launay
- Medical Oncology Department, Paoli Calmettes Institute, Marseille, France
| | - Ludovic Evesque
- Pole of Medicine/Gastro-enterology, Antoine Lacassagne Centre, Nice, France
| | - Angélique Vienot
- Medical Oncology Department, Jean Minjoz Hospital, Besançon, France
| | - Hervé Perrier
- Oncology Department, Saint Joseph Hospital, Marseille, France
| | - Marine Jary
- Oncology Department, Nord Franche Comté Hospital, Montbéliard, France
| | - Yves Rinaldi
- Hepato-gastroenterology and Digestive Oncology Department, European Hospital of Marseille, Marseille, France
| | - Clélia Coutzac
- Medical Oncology Department, Centre Leon Bérard, University Lyon I, Lyon, France
| | - Jean Baptiste Bachet
- Hepato-gastroenterology and Digestive Oncology Department, Pitié-Salpêtrière Hospital, Paris, France
| | - Cindy Neuzillet
- Medical Oncology Department, Curie Institute, Saint Cloud, France
| | - Nicolas Williet
- Hepato-gastroenterology Department, University Institute of Cancerology and Hematology of Saint-Etienne (ICHUSE), Targeting Research Unit in Oncology at the University Hospital of Saint-Etienne (URCAS), Saint-Priest-en-Jarez, France
| | - Romain Desgrippes
- Hepato-Gastroenterology and Digestive Oncology Department, Broussais Hospital, Saint-Malo, France
| | | | - Thomas Aparicio
- Gastroenterology and Digestive Cancerology Department, Saint Louis Hospital, Paris, France
| | - Annie Peytier
- Hepato-Gastroenterology Department, Hospital Centre of Bayeux, Bayeux, France
| | - Thierry Lecomte
- Hepato-gastroenterology and Digestive Oncology Department, CHU Tours, Tours, France
- Department of Nutrition, Growth and Cancer, INSERM UMR1069, University of Tours, Tours, France
| | - Gaël S Roth
- Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Institute for Advanced Biosciences, CNRS UMR 5309-INSERM U1209, Université Grenoble Alpes, Grenoble, France
| | - Anne Thirot-Bidault
- Institute of Cancerology Paris Sud-Private Hospital of Antony, Antony, France
| | | | - Olivier Bouché
- Hepatogastroenterology and Digestive Cancerology Department, CHU Robert Debré, Reims, France
| | | |
Collapse
|
3
|
Singh M, Jana BK, Pal P, Singha I, Rajkumari A, Chowrasia P, Nath V, Mazumder B. Nanoparticles in pancreatic cancer therapy: a detailed and elaborated review on patent literature. Expert Opin Ther Pat 2023; 33:681-699. [PMID: 37991186 DOI: 10.1080/13543776.2023.2287520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/21/2023] [Indexed: 11/23/2023]
Abstract
INTRODUCTION Nanotechnology may open up new avenues for overcoming the challenges of pancreatic cancer therapy as a broad arsenal of anticancer medicines fail to realize their full therapeutic potential in pancreatic ductal adenocarcinoma due to the formation of multiple resistance mechanisms inside the tumor. Many studies have reported the successful use of various nano formulations in pancreatic cancer therapy. AREAS COVERED This review covers all the major nanotechnology-based patent litrature available on renowned patent data bases like Patentscope and Espacenet, through the time period of 2007-2022. This is an entirely patent centric review, and it includes both clinical and non-clinical data available on nanotechnology-based therapeutics and diagnostic tools for pancreatic cancer. EXPERT OPINION For the sake of understanding, the patents are categorized under various formulation-specific heads like metallic/non-metallic nanoparticles, polymeric nanoparticles, liposomes, carbon nanotubes, protein nanoparticles and liposomes. This distinguishes one specific nanoparticle type from another and makes this review a one-of-a-kind comprehensive patent compilation that has not been reported so far in the history of nanotechnological formulations in pancreatic cancer.
Collapse
Affiliation(s)
- Mohini Singh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Bani Kumar Jana
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Paulami Pal
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Ishita Singha
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Ananya Rajkumari
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Pinky Chowrasia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Venessa Nath
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Bhaskar Mazumder
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| |
Collapse
|
4
|
Periyasamy L, Murugantham B, Muthusami S. Plumbagin binds to epidermal growth factor receptor and mitigate the effects of epidermal growth factor micro-environment in PANC-1 cells. Med Oncol 2023; 40:184. [PMID: 37209241 DOI: 10.1007/s12032-023-02048-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/05/2023] [Indexed: 05/22/2023]
Abstract
A sustained increase in the mortality of pancreatic cancer (PC) and sudden metastasis-related mortality is a cause for concern. Aberrant expression of epidermal growth factor (EGF) receptor (EGFR) is noted in several cases of PC metastasis. The present study is aimed at analyzing the expression of EGFR in PC and its relevance to the progression of PC. Despite the number of studies that have shown the benefits of plumbagin on PC cells, its role on cancer stem cells remains largely unknown. To this end, the study used an EGF micro-environment to make cancer stem cells in vitro and ascertained the role of plumbagin in mitigating the actions of EGF. The kaplan-meier (KM) plot indicated reduced overall survival (OS) analysis in PC patients with high EGFR than low EGFR expression. Plumbagin pre-treatment significantly prevented EGF-induced survival, epithelial-to-mesenchymal transition (EMT), clonogenesis, migration, matrix metalloproteinase -2 (MMP-2) gene expression and its secretion, and matrix protein hyaluron production in PANC-1 cells. The computational studies indicate the greater affinity of plumbagin with different domains of EGFR than gefitinib. Several hallmarks of resistance and migration due to EGF are effectively attenuated by plumbagin. Collectively, these results warrant investigating the actions of plumbagin in a pre-clinical study to substantiate these findings.
Collapse
Affiliation(s)
- Loganayaki Periyasamy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India
| | - Bharathi Murugantham
- Karpagam Cancer Research Centre, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India
| | - Sridhar Muthusami
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India.
- Karpagam Cancer Research Centre, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India.
| |
Collapse
|
5
|
Palma AM, Vudatha V, Peixoto ML, Madan E. Tumor heterogeneity: An oncogenic driver of PDAC progression and therapy resistance under stress conditions. Adv Cancer Res 2023; 159:203-249. [PMID: 37268397 DOI: 10.1016/bs.acr.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a clinically challenging disease usually diagnosed at advanced or metastasized stage. By this year end, there are an expected increase in 62,210 new cases and 49,830 deaths in the United States, with 90% corresponding to PDAC subtype alone. Despite advances in cancer therapy, one of the major challenges combating PDAC remains tumor heterogeneity between PDAC patients and within the primary and metastatic lesions of the same patient. This review describes the PDAC subtypes based on the genomic, transcriptional, epigenetic, and metabolic signatures observed among patients and within individual tumors. Recent studies in tumor biology suggest PDAC heterogeneity as a major driver of disease progression under conditions of stress including hypoxia and nutrient deprivation, leading to metabolic reprogramming. We therefore advance our understanding in identifying the underlying mechanisms that interfere with the crosstalk between the extracellular matrix components and tumor cells that define the mechanics of tumor growth and metastasis. The bilateral interaction between the heterogeneous tumor microenvironment and PDAC cells serves as another important contributor that characterizes the tumor-promoting or tumor-suppressing phenotypes providing an opportunity for an effective treatment regime. Furthermore, we highlight the dynamic reciprocating interplay between the stromal and immune cells that impact immune surveillance or immune evasion response and contribute towards a complex process of tumorigenesis. In summary, the review encapsulates the existing knowledge of the currently applied treatments for PDAC with emphasis on tumor heterogeneity, manifesting at multiple levels, impacting disease progression and therapy resistance under stress.
Collapse
Affiliation(s)
| | - Vignesh Vudatha
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | | | - Esha Madan
- Champalimaud Centre for the Unknown, Lisbon, Portugal; Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.
| |
Collapse
|
6
|
Xu B, Shi J, Lu W, Wu Y, Dong X. Elderly Pancreatic Adenocarcinoma Cancer Patients Could Benefit From Postoperative Chemotherapy. Pancreas 2023; 52:e37-e44. [PMID: 37165831 PMCID: PMC10317297 DOI: 10.1097/mpa.0000000000002214] [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: 12/19/2021] [Accepted: 02/15/2023] [Indexed: 05/12/2023]
Abstract
OBJECTIVES The study aim to investigate whether elderly patients with resectable pancreatic ductal adenocarcinoma (PDAC) could benefit from postoperative chemotherapy. METHODS This study selects the data of PDAC patients who were diagnosed between 2004 and 2014 from the Surveillance, Epidemiology, and End Results program. Median overall survival (mOS) is determined by Kaplan-Meier survival curves. Multivariate logistic regression analysis and hazard ratio are employed to assess the association among potential prognostic factors. Propensity score matching evaluation is used to reduce bias. RESULTS In total, there are 11,865 PDAC patients selected from the Surveillance, Epidemiology, and End Results database. Elderly PDAC patients have poor prognoses compared with younger (mOS, 15 vs 21 months). The possible reason might be that the elderly patients are less likely to receive postoperative chemotherapy. After propensity score matching, it is found that, for those who receive postoperative chemotherapy, although the mOS of older group is not as good as that of the younger group (mOS, 20 vs 23 months; 18-month survival rate: 53.4% vs 61.3%), the mOS of older group prolonged by postoperative chemotherapy is similar to that of younger group (9 vs 9 months). CONCLUSIONS Elderly PDAC patients (≥70 years) might benefit from the currently used postoperative chemotherapy regimens.
Collapse
Affiliation(s)
- Bin Xu
- From the Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinbo Shi
- From the Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenjie Lu
- From the Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yulian Wu
- From the Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Dong
- From the Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
7
|
Singh M, Pal P, Dutta RS, Marbaniang D, Ray S, Mazumder B. Nanodiamond Mediated Molecular Targeting in Pancreatic Ductal Adenocarcinoma: Disrupting the Tumor-stromal Cross-talk, Next Hope on the Horizon? Curr Cancer Drug Targets 2023; 23:620-633. [PMID: 36843367 DOI: 10.2174/1568009623666230227120837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 02/28/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the foremost causes of cancer-related morbidities worldwide. Novel nanotechnology-backed drug delivery stratagems, including molecular targeting of the chemotherapeutic payload, have been considered. However, no quantum leap in the gross survival rate of patients with PDAC has been realized. One of the predominant causes behind this is tumor desmoplasia, a dense and heterogenous stromal extracellular matrix of the tumor, aptly termed tumor microenvironment (TME). It plays a pivotal role in the tumor pathogenesis of PDAC as it occupies most of the tumor mass, making PDAC one of the most stromal-rich cancers. The complex crosstalk between the tumor and dynamic components of the TME impacts tumor progression and poses a potential barrier to drug delivery. Understanding and deciphering the complex cascade of tumorstromal interactions are the need of the hour so that we can develop neoteric nano-carriers to disrupt the stroma and target the tumor. Nanodiamonds (NDs), due to their unique surface characteristics, have emerged as a promising nano delivery system in various pre-clinical cancer models and have the potential to deliver the chemotherapeutic payload by moving beyond the dynamic tumor-stromal barrier. It can be the next revolution in nanoparticle-mediated pancreatic cancer targeting.
Collapse
Affiliation(s)
- Mohini Singh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Paulami Pal
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Rajat Subhra Dutta
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Daphisha Marbaniang
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Subhabrata Ray
- Dr. B.C. Roy College of Pharmacy & AHS, Durgapur, WB, India
| | - Bhaskar Mazumder
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| |
Collapse
|
8
|
Kindler HL, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, Park JO, Hochhauser D, Arnold D, Oh DY, Reinacher-Schick A, Tortora G, Algül H, O'Reilly EM, Bordia S, McGuinness D, Cui K, Locker GY, Golan T. Overall Survival Results From the POLO Trial: A Phase III Study of Active Maintenance Olaparib Versus Placebo for Germline BRCA-Mutated Metastatic Pancreatic Cancer. J Clin Oncol 2022; 40:3929-3939. [PMID: 35834777 PMCID: PMC10476841 DOI: 10.1200/jco.21.01604] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 03/21/2022] [Accepted: 06/02/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE The phase III POLO study demonstrated significant progression-free survival (PFS) benefit for active olaparib maintenance therapy versus placebo for patients with metastatic pancreatic adenocarcinoma and a germline BRCA mutation. Here, we report the final analysis of overall survival (OS) and other secondary end points. PATIENTS AND METHODS Patients with a deleterious or suspected deleterious germline BRCA mutation whose disease had not progressed after ≥ 16 weeks of first-line platinum-based chemotherapy were randomly assigned 3:2 to active maintenance olaparib (300 mg twice daily) or placebo. The primary end point was PFS; secondary end points included OS, time to second disease progression or death, time to first and second subsequent cancer therapies or death, time to discontinuation of study treatment or death, and safety and tolerability. RESULTS In total, 154 patients were randomly assigned (olaparib, n = 92; placebo, n = 62). No statistically significant OS benefit was observed (median 19.0 v 19.2 months; hazard ratio [HR], 0.83; 95% CI, 0.56 to 1.22; P = .3487). Kaplan-Meier OS curves separated at approximately 24 months, and the estimated 3-year survival after random assignment was 33.9% versus 17.8%, respectively. Median time to first subsequent cancer therapy or death (HR, 0.44; 95% CI, 0.30 to 0.66; P < .0001), time to second subsequent cancer therapy or death (HR, 0.61; 95% CI, 0.42 to 0.89; P = .0111), and time to discontinuation of study treatment or death (HR, 0.43; 95% CI, 0.29 to 0.63; P < .0001) significantly favored olaparib. The HR for second disease progression or death favored olaparib without reaching statistical significance (HR, 0.66; 95% CI, 0.43 to 1.02; P = .0613). Olaparib was well tolerated with no new safety signals. CONCLUSION Although no statistically significant OS benefit was observed, the HR numerically favored olaparib, which also conferred clinically meaningful benefits including increased time off chemotherapy and long-term survival in a subset of patients.
Collapse
Affiliation(s)
| | - Pascal Hammel
- Paul Brousse Hospital (AP-HP), University Paris-Saclay, Villejuif, France
| | - Michele Reni
- IRCCS Ospedale, San Raffaele Scientific Institute, Vita e Salute University, Milan, Italy
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg and KU Leuven, Leuven, Belgium
| | - Teresa Macarulla
- Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Joon Oh Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Dirk Arnold
- Asklepios Tumorzentrum Hamburg AK Altona, Hamburg, Germany
| | - Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | | | - Giampaolo Tortora
- Fondazione Policlinico A Gemelli IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Hana Algül
- Klinikum rechts der Isar, Comprehensive Cancer Center Munich TUM, Technische Universität München, Munich, Germany
| | | | | | | | | | | | - Talia Golan
- The Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
9
|
Principe DR, Aissa AF, Kumar S, Pham TND, Underwood PW, Nair R, Ke R, Rana B, Trevino JG, Munshi HG, Benevolenskaya EV, Rana A. Calcium channel blockers potentiate gemcitabine chemotherapy in pancreatic cancer. Proc Natl Acad Sci U S A 2022; 119:e2200143119. [PMID: 35476525 PMCID: PMC9170157 DOI: 10.1073/pnas.2200143119] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/19/2022] [Indexed: 12/15/2022] Open
Abstract
There is currently no effective treatment for pancreatic ductal adenocarcinoma (PDAC). While palliative chemotherapy offers a survival benefit to most patients, nearly all will eventually progress on treatment and long-term survivability remains poor. Given the lack of subsequent line treatment options, in this study, we sought to identify novel strategies to prevent, delay, or overcome resistance to gemcitabine, one of the most widely used medications in PDAC. Using a combination of single-cell RNA sequencing and high-throughput proteomic analysis, we identified a subset of gemcitabine-resistant tumor cells enriched for calcium/calmodulin signaling. Pharmacologic inhibition of calcium-dependent calmodulin activation led to the rapid loss of drug-resistant phenotypes in vitro, which additional single-cell RNA sequencing identified was due to impaired activation of the RAS/ERK signaling pathway. Consistent with these observations, calcium chelation or depletion of calcium in the culture media also impaired ERK activation in gemcitabine-resistant cells, and restored therapeutic responses to gemcitabine in vitro. We observed similar results using calcium channel blockers (CCBs) such as amlodipine, which inhibited prosurvival ERK signaling in vitro and markedly enhanced therapeutic responses to gemcitabine in both orthotopic xenografts and transgenic models of PDAC. Combined, these results offer insight into a potential means of gemcitabine resistance and suggest that select CCBs may provide a clinical benefit to PDAC patients receiving gemcitabine-based chemotherapy.
Collapse
Affiliation(s)
- Daniel R. Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL 60612
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60612
| | - Alexandre F. Aissa
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60612
| | - Sandeep Kumar
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Thao N. D. Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL 60611
| | - Patrick W. Underwood
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32611
| | - Rakesh Nair
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Rong Ke
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Basabi Rana
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Jose G. Trevino
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University, Richmond, VA 23284
| | - Hidayatullah G. Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL 60611
- Jesse Brown VA Medical Center, Chicago, IL 60612
| | | | - Ajay Rana
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
- Jesse Brown VA Medical Center, Chicago, IL 60612
| |
Collapse
|
10
|
Kamposioras K, Papaxoinis G, Dawood M, Appleyard J, Collinson F, Lamarca A, Ahmad U, Hubner RA, Wright F, Pihlak R, Damyanova I, Razzaq B, Valle JW, McNamara MG, Anthoney A. Markers of tumor inflammation as prognostic factors for overall survival in patients with advanced pancreatic cancer receiving first-line FOLFIRINOX chemotherapy. Acta Oncol 2022; 61:583-590. [PMID: 35392758 DOI: 10.1080/0284186x.2022.2053198] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 03/08/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Identifying pretreatment blood markers that distinguish prognostic groups of patients with advanced pancreatic ductal adenocarcinoma (PDAC) under first-line FOLFIRINOX chemotherapy has the potential to improve management of this condition. Aim of this study was to determine the prognostic utility of a range of pretreatment, inflammation-related, blood cell markers in this group of patients. MATERIAL AND METHODS Data from a training cohort were analyzed to identify potential pretreatment blood markers correlating to survival outcomes. The most informative markers were further analyzed in a validation cohort comprised patients from a geographically separate cancer center undergoing the same treatment. RESULTS A total of 138 consecutive patients receiving FOLFIRINOX chemotherapy between 2010 and 2019, constituted the training cohort. Neutrophil/lymphocyte (NLR), monocyte/lymphocyte (MLR), and platelet/lymphocyte ratio (PLR) as well as the systemic inflammatory response index (SIRI) and CA19.9 showed prognostic significance in addition to tumor stage. A pretreatment SIRI score cutoff of 2.35 differentiated between a poor prognostic group with median overall survival (mOS) 5.1 months and a better prognostic group, mOS 12.5 months. SIRI ≤/> 2.35 was predictive of mOS in patients with locally advanced and metastatic PDAC. SIRI was confirmed as a prognostic marker in a validation cohort of 67 patients with mOS of 13.4 months and 6.3 months for those with SIRI ≤ 2.35 and >2.35, respectively. Additional analysis revealed baseline SIRI as being prognostic within additional subgroups of patients in both cohorts. CONCLUSIONS This large, retrospective, analysis of real-world patients receiving first-line FOLFIRINOX chemotherapy for advanced PDAC has identified the pretreatment blood SIRI as a strong prognostic marker for survival. This will allow better counseling of patients with regards to the benefits of treatment, improved stratification within clinical trials, and potentially identify groups of patients for novel therapy trials as first-line treatment.
Collapse
Affiliation(s)
| | - George Papaxoinis
- Second Department of Oncology, Agios Savvas Anticancer Hospital, Athens, Greece
| | - Mohamed Dawood
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Jordan Appleyard
- Barts and The London School of Medicine and Dentistry, London, UK
| | - Fiona Collinson
- Department of Medical Oncology, Leeds Institute for Medical Research, St James' Institute of Oncology, St James' University Hospital, University of Leeds, Leeds, UK
| | - Angela Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust/Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Usman Ahmad
- Department of Medical Oncology, The Mid Yorkshire Hospitals NHS Trust, Wakefield, UK
| | - Richard A Hubner
- Department of Medical Oncology, The Christie NHS Foundation Trust/Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Francesca Wright
- Department of Medical Oncology, Leeds Institute for Medical Research, St James' Institute of Oncology, St James' University Hospital, University of Leeds, Leeds, UK
| | - Rille Pihlak
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Iva Damyanova
- Department of Medical Oncology, The Mid Yorkshire Hospitals NHS Trust, Wakefield, UK
| | - Bilal Razzaq
- Department of Medical Oncology, The Mid Yorkshire Hospitals NHS Trust, Wakefield, UK
| | - Juan W Valle
- Division of Cancer Sciences, University of Manchester/The Christie NHS Foundation Trust, Manchester, UK
| | - Mairéad G McNamara
- Division of Cancer Sciences, University of Manchester/The Christie NHS Foundation Trust, Manchester, UK
| | - Alan Anthoney
- Department of Medical Oncology, Leeds Institute for Medical Research, St James' Institute of Oncology, St James' University Hospital, University of Leeds, Leeds, UK
| |
Collapse
|
11
|
Principe DR. Precision Medicine for BRCA/PALB2-Mutated Pancreatic Cancer and Emerging Strategies to Improve Therapeutic Responses to PARP Inhibition. Cancers (Basel) 2022; 14:cancers14040897. [PMID: 35205643 PMCID: PMC8869830 DOI: 10.3390/cancers14040897] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/01/2022] [Accepted: 02/08/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary For the small subset of pancreatic ductal adenocarcinoma (PDAC) patients with loss-of-function mutations to BRCA1/2 or PALB2, both first-line and maintenance therapy differs significantly. These mutations confer a loss of double-strand break DNA homologous recombination (HR), substantially altering drug sensitivities. In this review, we discuss the current treatment guidelines for PDAC tumors deficient in HR, as well as newly emerging strategies to improve drug responses in this population. We also highlight additional patient populations in which these strategies may also be effective, and novel strategies aiming to confer similar drug sensitivity to tumors proficient in HR repair. Abstract Pancreatic cancer is projected to become the second leading cause of cancer-related death by 2030. As patients typically present with advanced disease and show poor responses to broad-spectrum chemotherapy, overall survival remains a dismal 10%. This underscores an urgent clinical need to identify new therapeutic approaches for PDAC patients. Precision medicine is now the standard of care for several difficult-to-treat cancer histologies. Such approaches involve the identification of a clinically actionable molecular feature, which is matched to an appropriate targeted therapy. Selective poly (ADP-ribose) polymerase (PARP) inhibitors such as Niraparib, Olaparib, Talazoparib, Rucaparib, and Veliparib are now approved for several cancers with loss of high-fidelity double-strand break homologous recombination (HR), namely those with deleterious mutations to BRCA1/2, PALB2, and other functionally related genes. Recent evidence suggests that the presence of such mutations in pancreatic ductal adenocarcinoma (PDAC), the most common and lethal pancreatic cancer histotype, significantly alters drug responses both with respect to first-line chemotherapy and maintenance therapy. In this review, we discuss the current treatment paradigm for PDAC tumors with confirmed deficits in double-strand break HR, as well as emerging strategies to both improve responses to PARP inhibition in HR-deficient PDAC and confer sensitivity to tumors proficient in HR repair.
Collapse
Affiliation(s)
- Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
12
|
Zhang W, Xu L, Che X. Nomogram for Predicting the Prognoses of Patients With Pancreatic Head Cancer After Pancreaticoduodenectomy: A Population-Based Study on SEER Data. Front Oncol 2021; 11:766071. [PMID: 34858844 PMCID: PMC8631716 DOI: 10.3389/fonc.2021.766071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/18/2021] [Indexed: 01/21/2023] Open
Abstract
Objective In this study, we retrieved the data available in the Surveillance, Epidemiology, and End Results database to identify the prognostic factors for patients with pancreatic head cancer who had undergone pancreaticoduodenectomy and developed a prediction model for clinical reference. Methods We screened the data between 1973 and 2015. Propensity score matching (PSM) was used to control for the confounding factors. Kaplan-Meier (log-rank test) curves were used to compare the survival rates. A nomogram was established using multifactorial Cox regression. Results In total, 4099 patients were identified. Their median survival was 22 months, with 74.2%, 36.5%, and 26.2% survival after 1, 3, and 5 years, respectively. The median cancer-specific survival was 24.0 months, with 71.1%, 32.6%, and 21.9% survival after 1, 3, and 5 years, respectively. The results of the Cox proportional risk regression showed that age, insurance status, gender, histological type, degree of tissue differentiation, T and N stages, tumor size, extent of regional lymph node dissection, and postoperative radiotherapy or chemotherapy are independent factors affecting prognosis. PSM was used twice to eliminate any bias from the unbalanced covariates in the raw data. After PSM, the patients who had received postoperative radiotherapy were found to have a better survival prognosis and disease-specific survival prognosis than those who had not received radiotherapy [HR = 0.809, 95% CI (0.731–0.894), P < 0.001 and HR = 0.814, 95% CI (0.732–0.904), P < 0.001; respectively]. A similar result was observed for the patients who had received postoperative chemotherapy versus those who had not [HR = 0.703, 95% CI (0.633–0.78), P < 0.001 and HR = 0.736, 95% CI (0.658–0.822), P < 0.001, for survival and disease-specific survival prognoses, respectively]. Finally, the β coefficients of the Cox proportional risk regression were used to establish a nomogram. Conclusion Age, insurance status, gender, histological type, degree of differentiation, T and N stages, tumor size, regional lymph node dissection, and postoperative radiotherapy or chemotherapy are factors affecting the prognosis in pancreatic head cancer after pancreaticoduodenectomy. Postoperative radiotherapy and chemotherapy can improve patient survival. These still need to be further validated in the future.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China.,Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Xu
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Xu Che
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China.,Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Inhibition of JAK2/STAT3 signaling pathway by panaxadiol limits the progression of pancreatic cancer. Aging (Albany NY) 2021; 13:22830-22842. [PMID: 34623971 PMCID: PMC8544303 DOI: 10.18632/aging.203575] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/18/2021] [Indexed: 11/25/2022]
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related death with the characteristics of chemoresistance and early metastasis. Panaxadiol, a triterpenoid saponin extracted from the roots of American ginseng, has been proved to display anti-tumor activity in colon cancer. In this study, we found panaxadiol significantly inhibited proliferation, and induced apoptosis in human pancreatic cancer cell lines PANC-1 and Patu8988 in a dose-dependent manner. Furthermore, the expression of apoptosis-related proteins (Bax, Bcl2, Cleaved-caspase3) was detected via western blot and immunofluorescence staining. In addition, panaxadiol was also found to inhibit the migration of pancreatic cancer cells by wound healing and transwell assays. In vivo, the growth of xenograft pancreatic cancer models was also notably suppressed by panaxadiol compared to the control group. Moreover, the down-regulation of JAK2-STAT3 signaling pathway was responsible for the underlying pro-apoptosis mechanism of panaxadiol, and this result was in good agreement with molecular docking analysis between panaxadiol and STAT3. In conclusion, our work comprehensively explored the anti-tumor ability in PANC-1 and Patu8988 cells of panaxadiol and provided a potential choice for the clinical treatment of pancreatic cancer patients.
Collapse
|
14
|
Nab-paclitaxel plus S-1 with or without PD-1 inhibitor in pancreatic ductal adenocarcinoma with only hepatic metastases: a retrospective cohort study. Langenbecks Arch Surg 2021; 407:633-643. [PMID: 34518900 DOI: 10.1007/s00423-021-02321-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/27/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE The evidence regarding programmed cell death 1 (PD-1) inhibitors on pancreatic ductal adenocarcinoma (PDAC) with metastases remains controversial. This study aimed to assess the efficacy and safety of Nab-paclitaxel plus S1 (NPS) with or without Sintilimab, a PD-1 inhibitor, in patients with PDAC with only hepatic metastases (mPDAC). METHODS Untreated mPDAC patients who received NPS with (the combination group) or without Sintilimab (the NPS group) were retrospectively studied. Surgery was considered when the pancreatic tumor became resectable or borderline resectable on radiological examinations, and with complete metabolic response of liver metastases. RESULTS Between October 2017 and February 2020, 32 patients were in the combination group and 34 patients in the NPS group. Successful salvage resection was achieved in 17 (25.8%) patients after tumor-downstaging (combination 12 vs. NPS 5, P = 0.03). The median overall survival (OS) was 16.8 months in the combination group and 10.0 months in the NPS group (P = 0.002). Remarkable OS benefit was observed in patients with decline in CA19-9 of ≥ 50% (16.0 vs. 6.5, P = 0.003), reduction in 18F-fluorodeoxyglucose uptake of primary tumor of ≥ 50% (16.5 vs. 10.0, P < 0. 001) and after salvage resection (20.1 vs. 11.0, P < 0. 001). No significant difference in Grade 3 or higher adverse events were seen between the two groups (40.6% vs. 32.4%, P = 0.49). CONCLUSIONS Despite the inherent biases of this retrospective study, the addition of Sintilimab significantly improved salvage resection rates and OS compared with the NPS regimen and had a favorable safety profile in treatment naïve mPDAC patients.
Collapse
|
15
|
Principe DR, Underwood PW, Korc M, Trevino JG, Munshi HG, Rana A. The Current Treatment Paradigm for Pancreatic Ductal Adenocarcinoma and Barriers to Therapeutic Efficacy. Front Oncol 2021; 11:688377. [PMID: 34336673 PMCID: PMC8319847 DOI: 10.3389/fonc.2021.688377] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, with a median survival time of 10-12 months. Clinically, these poor outcomes are attributed to several factors, including late stage at the time of diagnosis impeding resectability, as well as multi-drug resistance. Despite the high prevalence of drug-resistant phenotypes, nearly all patients are offered chemotherapy leading to modest improvements in postoperative survival. However, chemotherapy is all too often associated with toxicity, and many patients elect for palliative care. In cases of inoperable disease, cytotoxic therapies are less efficacious but still carry the same risk of serious adverse effects, and clinical outcomes remain particularly poor. Here we discuss the current state of pancreatic cancer therapy, both surgical and medical, and emerging factors limiting the efficacy of both. Combined, this review highlights an unmet clinical need to improve our understanding of the mechanisms underlying the poor therapeutic responses seen in patients with PDAC, in hopes of increasing drug efficacy, extending patient survival, and improving quality of life.
Collapse
Affiliation(s)
- Daniel R. Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, United States
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Murray Korc
- Department of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Jose G. Trevino
- Department of Surgery, Division of Surgical Oncology, Virginia Commonwealth University, Richmond, VA, United States
| | - Hidayatullah G. Munshi
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Jesse Brown VA Medical Center, Chicago, IL, United States
| | - Ajay Rana
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
- Jesse Brown VA Medical Center, Chicago, IL, United States
| |
Collapse
|
16
|
Wang S, Zheng Y, Yang F, Zhu L, Zhu XQ, Wang ZF, Wu XL, Zhou CH, Yan JY, Hu BY, Kong B, Fu DL, Bruns C, Zhao Y, Qin LX, Dong QZ. The molecular biology of pancreatic adenocarcinoma: translational challenges and clinical perspectives. Signal Transduct Target Ther 2021; 6:249. [PMID: 34219130 PMCID: PMC8255319 DOI: 10.1038/s41392-021-00659-4] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is an increasingly common cause of cancer mortality with a tight correspondence between disease mortality and incidence. Furthermore, it is usually diagnosed at an advanced stage with a very dismal prognosis. Due to the high heterogeneity, metabolic reprogramming, and dense stromal environment associated with pancreatic cancer, patients benefit little from current conventional therapy. Recent insight into the biology and genetics of pancreatic cancer has supported its molecular classification, thus expanding clinical therapeutic options. In this review, we summarize how the biological features of pancreatic cancer and its metabolic reprogramming as well as the tumor microenvironment regulate its development and progression. We further discuss potential biomarkers for pancreatic cancer diagnosis, prediction, and surveillance based on novel liquid biopsies. We also outline recent advances in defining pancreatic cancer subtypes and subtype-specific therapeutic responses and current preclinical therapeutic models. Finally, we discuss prospects and challenges in the clinical development of pancreatic cancer therapeutics.
Collapse
Affiliation(s)
- Shun Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Yan Zheng
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Feng Yang
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Le Zhu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xiao-Qiang Zhu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhe-Fang Wang
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Xiao-Lin Wu
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Cheng-Hui Zhou
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Jia-Yan Yan
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bei-Yuan Hu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Bo Kong
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - De-Liang Fu
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Christiane Bruns
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Yue Zhao
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany.
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.
| | - Qiong-Zhu Dong
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.
- Key laboratory of whole-period monitoring and precise intervention of digestive cancer, Shanghai Municipal Health Commission (SMHC), Shanghai, China.
| |
Collapse
|
17
|
Xie D, Qian B, Yang J, Peng X, Li Y, Hu T, Lu S, Chen X, Han Y. Can Elderly Patients With Pancreatic Cancer Gain Survival Advantages Through More Radical Surgeries? A SEER-Based Analysis. Front Oncol 2020; 10:598048. [PMID: 33194764 PMCID: PMC7660699 DOI: 10.3389/fonc.2020.598048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 10/06/2020] [Indexed: 11/13/2022] Open
Abstract
Background and Aims In recent years, the best treatment method for pancreatic cancer in elderly patients has remained controversial. Surgery is the main treatment modality for pancreatic cancer. This study aimed to determine whether elderly patients with pancreatic cancer can gain survival advantages through more active and radical surgical treatment and evaluate the best treatment method and potential prognostic factors. Methods From the Surveillance, Epidemiology, and End Results program (SEER) database, 10,557 elderly patients (aged ≥65 years) with pancreatic cancer were included as Cohort 1, and Propensity Score Matching (PSM) evaluation was performed to generate Cohort 2 (424 pairs). Overall Survival (OS) and Cause-Specific Survival (CSS) were determined using Kaplan-Meier survival curves, and differences were assessed using the Log-rank test. Multivariate logistic regression analysis and the forest plot of hazard ratio (HR) was made to assess the association between potential prognostic factors, including surgery and different surgical methods, and survival in elderly patients. Results We identified 10,557 eligible patients with pancreatic cancer, who formed Cohort 1. The total OS and CSS in the surgery group were significantly higher than those in the non-surgery group (P < 0.001). Age, stage (AJCC 8th), grade, lymph node metastasis, radiation, chemotherapy, and surgical methods were independent factors affecting the prognosis of elderly patients. In Cohort 2, Total pancreatectomy (Total PT) had the lowest risk ratio (HR = 0.31, P < 0.001) and longest median CSS (18.000 months), while Extension Total pancreatectomy (Ex-Total PT, HR = 0.34, P < 0.001) showed the lower median CSS (17.000 months) and median OS (14.000 months). Partial pancreatectomy (Partial PT, HR = 0.46, P < 0.001) showed the lowest median CSS (13.000 months) and median OS (12.000 months), although they were still higher than the median CSS (6.000 months) and median OS (5.000 months) in the non-surgery group. Conclusions Based on the SEER database, surgical treatment is an independent prognostic factor in elderly patients with pancreatic cancer. Compared with other surgical methods, Total PT can offer elderly patients the best survival advantages. However, Ex-Total PT, a more radical method, does not seem to be the best treatment option for the survival and benefit of elderly patients.
Collapse
Affiliation(s)
- Danna Xie
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Baolin Qian
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jing Yang
- Department of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Xinya Peng
- Department of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yinghua Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Teng Hu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Simin Lu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaojing Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
18
|
Franco F, Camara JC, Martín-Valadés JI, López-Alfonso A, Marrupe D, Gutiérrez-Abad D, Martínez-Amores B, León A, Juez I, Pérez M, Royuela A, Ruiz-Casado A. Clinical outcomes of FOLFIRINOX and gemcitabine-nab paclitaxel for metastatic pancreatic cancer in the real world setting. Clin Transl Oncol 2020; 23:812-819. [PMID: 32857340 DOI: 10.1007/s12094-020-02473-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND/OBJECTIVES The incidence of pancreatic cancer is increasing in developed countries. The incorporation of new therapies, to the first-line treatment of patients with good performance status led to better survival in clinical trials. However, there is a wide variability in their use and some concerns about the treatment of elderly patients who were not included in the clinical trials. METHODS This is a retrospective multicenter study. Data from consecutive patients diagnosed with metastatic pancreatic cancer (mPC) treated with FOLFIRINOX (FFX) or gemcitabine plus nab-paclitaxel (GnP) were analysed to evaluate efficacy (overall survival-OS) and toxicity. RESULTS A total of 119 patients were included. 49.6% were treated with FFX and 50.4% with GNP in first-line. The median OS was 12 months with no statistically significant differences between both regimens (12.7 m for FFX vs 10.2 m for GnP). Elevated Ca 19.9 levels and neutrophil-lymphocyte ratio (NLR) increased the risk of death. Patients who received both regimens in first/second line had a median OS longer than 15 months whichever the sequence. 32 patients (27%) were older than 70-y. 54% patients received a second-line treatment, 56% in the FFX group and 44% in the GnP group. The median OS for patients older than 70 was 9.5 m versus 12.3 m for patients younger than 70. Progression of the disease was the cause of death in 67.6% of the patients. CONCLUSIONS In our setting, the use of FFX and GnP for treating mPC is quite similar, but superiority could not be demonstrated for any of the schemes in the first line. OS was determined by basal levels of Ca 19.9 and NLR. Patients receiving both regimens in first/second line whichever the sequence, exhibited the best survival rates. In our series, elderly patients had poorer survival rates.
Collapse
Affiliation(s)
- F Franco
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Majadahonda, Spain.
| | - J C Camara
- Department of Medical Oncology, Fundación Hospital Alcorcón, Alcorcón, Spain
| | | | - A López-Alfonso
- Department of Medical Oncology, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - D Marrupe
- Department of Medical Oncology, Hospital Universitario de Móstoles, Móstoles, Spain
| | - D Gutiérrez-Abad
- Department of Medical Oncology, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - B Martínez-Amores
- Department of Medical Oncology, Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | - A León
- Department of Medical Oncology, Fundación Jiménez Díaz, Madrid, Spain
| | - I Juez
- Department of Medical Oncology, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - M Pérez
- Department of Medical Oncology, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - A Royuela
- Department of Biostatistics, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - A Ruiz-Casado
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| |
Collapse
|
19
|
Kamal M, Wang XS, Shi Q, Mendoza T, Garcia-Gonzalez A, Bokhari RH, Cleeland CS, Fogelman DR. A Randomized, Placebo-Controlled, Double-Blind Study of Minocycline for Reducing the Symptom Burden Experienced by Patients With Advanced Pancreatic Cancer. J Pain Symptom Manage 2020; 59:1052-1058.e1. [PMID: 31982605 PMCID: PMC7249481 DOI: 10.1016/j.jpainsymman.2020.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 12/15/2022]
Abstract
CONTEXT Although it is well known that patients with advanced pancreatic cancer (PC) experience significant symptom burden, few strategies for effective symptom intervention are available for them. OBJECTIVES To investigate the efficacy of minocycline, an anti-inflammatory agent, for symptom reduction in patients with advanced PC. METHODS We conducted Phase II, randomized, and placebo-controlled trial to obtain preliminary estimates of the effects on symptom reduction with 100 mg of minocycline or placebo given twice a day. Eligible patients had diagnosed advanced PC and were scheduled for standard chemotherapy. Patient-reported symptoms were measured weekly during the eight-week trial using the MD Anderson Symptom Inventory (MDASI) module in patients with gastrointestinal cancer. The primary outcome measure was the area under the curve values of the five most severe symptoms in the two arms. RESULTS Of the 44 patients recruited, 31 (71%) were evaluable for the primary efficacy analysis, with 18 received minocycline and 13 placebo. Fatigue, pain, disturbed sleep, lack of appetite, and drowsiness were the most severe symptoms reported by both groups. No significant differences in area under the curve values over time between the study arms were found for the composite MDASI score or single-item scores of the five most severe MDASI items. No treatment-related deaths were reported, and no Grade 3-4 toxicities were observed. CONCLUSION Minocycline is safe for use in patients receiving treatment for PC. There is no observed symptom reduction with minocycline on the major symptom burden associated with advanced PC compared with placebo. Attrition because of rapid disease progression impacted the study significantly.
Collapse
Affiliation(s)
- Mona Kamal
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xin Shelley Wang
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Qiuling Shi
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tito Mendoza
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Araceli Garcia-Gonzalez
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Raza H Bokhari
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Charles S Cleeland
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David R Fogelman
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
20
|
Vreeland TJ, McAllister F, Javadi S, Prakash LR, Fogelman DR, Ho L, Varadhachary G, Aloia TA, Vauthey JN, Lee JE, Kim MP, Katz MHG, Tzeng CWD. Benefit of Gemcitabine/Nab-Paclitaxel Rescue of Patients With Borderline Resectable or Locally Advanced Pancreatic Adenocarcinoma After Early Failure of FOLFIRINOX. Pancreas 2019; 48:837-843. [PMID: 31210666 PMCID: PMC7309587 DOI: 10.1097/mpa.0000000000001345] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Neoadjuvant therapy (NT) is used for advanced pancreatic ductal adenocarcinoma (PDAC). No clear guidelines exist for switching therapies when patients do not respond to initial NT. We sought to characterize patients who underwent early switch from FOLFIRINOX to gemcitabine/nab-paclitaxel (GA) as NT for PDAC. METHODS We identified patients at a single institution switched from FOLFIRINOX to GA within the first 4 months of NT for PDAC during 2012-2017. We compared clinicopathologic data and oncologic outcomes. RESULTS Of 25 patients who met the criteria, 21 showed a serologic or radiographic response to GA; 11 (52%) reached resection. Responders had decreased carbohydrate antigen (CA) 19-9 levels from pretreatment to post-GA (P = 0.036). Resected responders had significantly decreased CA 19-9 comparing preswitch to post-GA (P = 0.048). The only predictor of GA response was prechemotherapy CA 19-9 of less than1000 U/mL (P = 0.021). Predictors of reaching resection were head/uncinate tumor (P = 0.010) and presenting stage lower than locally advanced (P = 0.041). CONCLUSIONS When patients do not respond to neoadjuvant FOLFIRINOX, early switch to GA should be considered. Future efforts should be directed toward identifying markers that will allow correct choice of initial therapy rather than attempting to rescue patients who respond poorly to first-line therapy.
Collapse
Affiliation(s)
- Timothy J Vreeland
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, Department of Gastrointestinal Medical Oncology, Clinical Cancer Genetics Program, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sanaz Javadi
- Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Laura R. Prakash
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - David R Fogelman
- Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Linus Ho
- Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gauri Varadhachary
- Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | |
Collapse
|
21
|
Matsumoto I, Kamei K, Omae K, Suzuki S, Matsuoka H, Mizuno N, Ozaka M, Ueno H, Kobayashi S, Uesugi K, Kobayashi M, Todaka A, Fukutomi A. FOLFIRINOX for locally advanced pancreatic cancer: Results and prognostic factors of subset analysis from a nation-wide multicenter observational study in Japan. Pancreatology 2019; 19:296-301. [PMID: 30638853 DOI: 10.1016/j.pan.2019.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/19/2018] [Accepted: 01/03/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND FOLFIRINOX (oxaliplatin, irinotecan, 5-fluorouracil, leucovorin) treatment significantly improved overall survival in the recent phase III study and became a standard therapy for metastatic pancreatic cancer. However, treatment for locally advanced pancreatic cancer is still controversial. We conducted subset analyses from a nation-wide multicenter observational study in Japan to evaluate the tolerability and efficacy of FOLFIRINOX in patients with locally advanced pancreatic cancer and to investigate independent prognostic factors with pre-treatment variables. METHODS The study included 66 patients with unresectable locally advanced pancreatic cancer from 27 institutions in Japan who received FOLFIRINOX as first-line treatment between December 20, 2013 and December 19, 2014 and surveyed until December 2015. RESULTS The median age was 63 with the Eastern Cooperative Oncology Group performance status of 0 or 1. Major Grade 3 or 4 adverse events included neutropenia (64%), leukopenia (33%), febrile neutropenia (15%), and diarrhea (15%). Severe adverse event occurred in 14 patients (11%) without fatal event. The median overall survival and progression-free survival times were 18.5 and 7.6 months, respectively. The objective response rate 15.2% and the disease control rate was 81.9%. A high modified Glasgow prognostic score (mGPS, ≥1) (95%CI 1.96-12.5) and female (95%CI 0.20-0.97) were identified as independent poor prognostic factors. CONCLUSIONS First-line FOLFIRINOX treatment for locally advanced pancreatic cancer seems to be effective with acceptable toxicities. A high mGPS may be associated with poor survival in patients with locally advanced pancreatic cancer who receive FOLFIRINOX. This study was registered at the UMIN Clinical Trials Registry (UMIN000014658).
Collapse
Affiliation(s)
- Ippei Matsumoto
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan.
| | - Keiko Kamei
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Katsuhiro Omae
- Clinical Research Center, Shizuoka Cancer Center, Shizuoka, Japan
| | - Shuhei Suzuki
- Department of Clinical Oncology, Yamagata University Hospital, Yamagata, Japan
| | | | - Nobumasa Mizuno
- Department of Gastroenterology, Aichi Cancer Center Hospital, Aichi, Japan
| | - Masato Ozaka
- Department of Gastroenterology, The Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hideki Ueno
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Satoshi Kobayashi
- Division of Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Kazuhiro Uesugi
- Department of Gastroenterology, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Marina Kobayashi
- Clinical Trial Promotion Section, Shizuoka Industrial Foundation Pharma Valley Center, Shizuoka, Japan
| | - Akiko Todaka
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Akira Fukutomi
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
22
|
Parkin A, Man J, Chou A, Nagrial AM, Samra J, Gill AJ, Timpson P, Pajic M. The Evolving Understanding of the Molecular and Therapeutic Landscape of Pancreatic Ductal Adenocarcinoma. Diseases 2018; 6:diseases6040103. [PMID: 30428574 PMCID: PMC6313363 DOI: 10.3390/diseases6040103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 12/18/2022] Open
Abstract
Pancreatic cancer is the third leading cause of cancer-related deaths, characterised by poor survival, marked molecular heterogeneity and high intrinsic and acquired chemoresistance. Only 10⁻20% of pancreatic cancer patients present with surgically resectable disease and even then, 80% die within 5 years. Our increasing understanding of the genomic heterogeneity of cancer suggests that the failure of definitive clinical trials to demonstrate efficacy in the majority of cases is likely due to the low proportion of responsive molecular subtypes. As a consequence, novel treatment strategies to approach this disease are urgently needed. Significant developments in the field of precision oncology have led to increasing molecular stratification of cancers into subtypes, where individual cancers are selected for optimal therapy depending on their molecular or genomic fingerprint. This review provides an overview of the current status of clinically used and emerging treatment strategies, and discusses the advances in and the potential for the implementation of precision medicine in this highly lethal malignancy, for which there are currently no curative systemic therapies.
Collapse
Affiliation(s)
- Ashleigh Parkin
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
| | - Jennifer Man
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
| | - Angela Chou
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
- University of Sydney, Sydney, NSW 2006, Australia.
| | - Adnan M Nagrial
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW 2145, Australia.
| | - Jaswinder Samra
- Department of Surgery, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia.
| | - Anthony J Gill
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
- University of Sydney, Sydney, NSW 2006, Australia.
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia.
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW 2065, Australia.
| | - Paul Timpson
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW 2010, Australia.
| | - Marina Pajic
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW 2010, Australia.
| |
Collapse
|
23
|
Olson JL, Bold RJ. Currently available first-line drug therapies for treating pancreatic cancer. Expert Opin Pharmacother 2018; 19:1927-1940. [PMID: 30325679 DOI: 10.1080/14656566.2018.1509954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Pancreatic adenocarcinoma is the 9th most common cancer in the United States and the 4th most common cause of cancer-related death given its poor prognosis. AREAS COVERED The authors have performed a literature search for pertinent published clinical trials, ongoing Phase 3 clinical trials, and current treatment guidelines using PubMed, Clinicaltrials.gov, and NCCN, ASCO, ESMO, and JPS websites. The review itself discusses landmark studies and ongoing research into the chemotherapy regimens recommended by each oncologic society. The authors also examine drugs that were promising but failed in Phase 3 trials and those currently being investigated. Finally, the authors provide their expert opinion on the subject and provide their future perspectives. EXPERT OPINION While advances in chemotherapy for pancreatic cancer have been limited in comparison to other cancers, there have been improvements in survival. Combination therapy and a goal of R0 resection are key elements to extend life. Novel agents directed at the unique properties of pancreatic cancer are promising.
Collapse
Affiliation(s)
- Jennifer L Olson
- a Division of Surgical Oncology , UC Davis Cancer Center , Sacramento , CA , USA
| | - Richard J Bold
- a Division of Surgical Oncology , UC Davis Cancer Center , Sacramento , CA , USA
| |
Collapse
|
24
|
|
25
|
Abstract
BACKGROUND There were many reports suggesting that different kinds of tumors can express B7-H4; however, the prognostic value in cancer was still unclearly. Therefore, we conducted a meta-analysis to investigate the relationship between overexpression of B7-H4 with the prognostic value in pancreatic cancer patients. MATERIALS AND METHODS The Pubmed, Embase, Cochrane Library, Ovid, Web of Science, and Chinese research database (including CBM, CNKI, and WAN FANG) were searched for related literature published until October 12, 2017. The pooled odds ratios (ORs) and/or pooled hazard ratios (HRs) for clinical pathological factors and overall survival (OS) were calculated and analyzed using Stata software. To assess whether an individual study had an impact on the result, sensitivity analysis was performed for all included individual studies using the fixed-effects model. Publication bias was evaluated using Egger's and Begg's tests. RESULTS Data from 6 observational studies including 442 patients were summarized in this meta-analysis, and each study was eligible for inclusion based on included and exclude criteria. The pooled results indicated that the B7-H4 overexpression could predict the presentation of lymph node metastasis (OR = 3.94, 95% CI: 1.22-12.66, P = .022), advanced TNM stage (T = the extent of the primary tumor, N = regional lymph nodes, M = distant metastases) (III+IV vs I+II; OR = 7.63, 95% CI: 2.46-23.66, P < .001), and the poor OS (HR = 3.00, 95%CI = 2.20-4.10, P < .001) in PC patients. CONCLUSIONS This study reveals that high expression of B7-H4 is an unfavorable prognostic factor for patients with pancreatic cancer. These results may guide the clinical management of this patient population.
Collapse
|