1
|
Kabir E, Azam F, Khan T, Yasmin H, Chowdhury N, Ahmed S, Sagar B, Tahrim N. Modeling Overall Survival in Patients With Pancreatic Cancer From a Pooled Analysis of Phase II Trials. Cancer Med 2024; 13:e70289. [PMID: 39387320 PMCID: PMC11465028 DOI: 10.1002/cam4.70289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 06/08/2024] [Accepted: 09/21/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND We evaluated the validity of surrogacy of progression-free survival (PFS) or time-to-progression (TTP) and overall response rate (ORR) in phase II trials of pancreatic ductal adenocarcinoma (PDAC). In addition, we explored the impact of predictive variables on overall survival (OS) and developed an optimal OS model. METHODS We analyzed 1867 clinical endpoint from 619 phase II PDAC trials with a systematic search from PubMed. Endpoint correlations were determined by Spearman's rank correlation. The assessed predictive factors included PFS/TTP, treatment size, therapy type, stage, and previous treatment. The relationship between predictors and OS was explored by a gamma generalized linear model (GLM) with a log-link function and compared with linear models. RESULTS The Spearman rank correlation coefficient between PFS/TTP and OS was 0.88 (95% confidence interval [CI] 0.85-0.89; p < 0.0001; n = 610) and between ORR and OS was 0.58 (0.52-0.64; p < 0.0001; n = 514). Model comparison favored the GLM model over the linear model, offering more accurate predictions for higher OS values. Consequently, PFS/TTP was the strongest predictor (pseudo-R2 = 0.75), with 1 added median PFS/TTP month associated with 13% (95% CI 13%-14%) increase in median OS. Subgroup analysis revealed that chemotherapy conferred significantly longer OS compared to targeted therapy in 1-Agent and 2-Agent trials, exhibiting a "very large" and "medium" effect size, respectively (rank biserial, rrb = 0.40 [95% CI 0.22-0.56] and rrb = 0.29 [0.16-0.41], both p < 0.0001), although inconsistent efficacy in 3-Agent trials (rrb = 0.12 [-0.07-0.30], p = 0.21). CONCLUSIONS PFS/TTP is a more reliable surrogate than ORR and a strong predictor of OS in phase II trials of pancreatic cancer. Moreover, gamma GLM (log-link function) is a robust tool for modeling positively skewed survival data with non-constant variance, thus can be applied to other cancers' OS data of such nature.
Collapse
Affiliation(s)
| | - Faruque Azam
- School of PharmacyBRAC UniversityDhakaBangladesh
| | | | | | | | | | | | | |
Collapse
|
2
|
Sharma R, Malviya R. Cancer Stem Cells in Carcinogenesis and Potential Role in Pancreatic Cancer. Curr Stem Cell Res Ther 2024; 19:1185-1194. [PMID: 37711007 DOI: 10.2174/1574888x19666230914103420] [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: 04/11/2023] [Revised: 07/14/2023] [Accepted: 08/11/2023] [Indexed: 09/16/2023]
Abstract
A poor prognosis is associated with pancreatic cancer because of resistance during treatment and early distant metastases. The discovery of cancer stem cells has opened up novel avenues for research into the biology and treatment of cancer. Many investigations have pointed out the role of these types of stem cells in the oncogenesis and progression of hematologic and solid malignancies, specifically. Due to the existence of cancer stem cells in the proliferation and preservation of pancreatic tumors, such malignancies could be difficult to eradicate using conventional treatment techniques like chemotherapy and radiotherapy. It is hypothesized that pancreatic malignancies originate from a limited population of aberrant cancer stem cells to promote carcinogenesis, tumour metastasis, and therapeutic resistance. This review examines the role of pancreatic cancer stem cells in this disease and their significance in carcinogenesis, as well as the signals which modulate them, and also examines the ongoing clinical studies that are now being conducted with pancreatic stem cells.
Collapse
Affiliation(s)
- Rishav Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
3
|
Chen EY, Kardosh A, Nabavizadeh N, Foster B, Mayo SC, Billingsley KG, Gilbert EW, Lanciault C, Grossberg A, Bensch KG, Maynard E, Anderson EC, Sheppard BC, Thomas CR, Lopez CD, Vaccaro GM. Phase 2 study of preoperative chemotherapy with nab-paclitaxel and gemcitabine followed by chemoradiation for borderline resectable or node-positive pancreatic ductal adenocarcinoma. Cancer Med 2023; 12:12986-12995. [PMID: 37132281 PMCID: PMC10315770 DOI: 10.1002/cam4.5971] [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: 12/28/2022] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Neoadjuvant treatment with nab-paclitaxel and gemcitabine for potentially operable pancreatic adenocarcinoma has not been well studied in a prospective interventional trial and could down-stage tumors to achieve negative surgical margins. METHODS A single-arm, open-label phase 2 trial (NCT02427841) enrolled patients with pancreatic adenocarcinoma deemed to be borderline resectable or clinically node-positive from March 17, 2016 to October 5, 2019. Patients received preoperative gemcitabine 1000 mg/m2 and nab-paclitaxel 125 mg/m2 on Days 1, 8, 15, every 28 days for two cycles followed by chemoradiation with 50.4 Gy intensity-modulated radiation over 28 fractions with concurrent fluoropyrimidine chemotherapy. After definitive resection, patients received four additional cycles of gemcitabine and nab-paclitaxel. The primary endpoint was R0 resection rate. Other endpoints included treatment completion rate, resection rate, radiographic response rate, survival, and adverse events. RESULTS Nineteen patients were enrolled, with the majority having head of pancreas primary tumors, both arterial and venous vasculature involvement, and clinically positive nodes on imaging. Among them, 11 (58%) underwent definitive resection and eight of 19 (42%) achieved R0 resection. Disease progression and functional decline were primary reasons for deferring surgical resection after neoadjuvant treatment. Pathologic near-complete response was observed in two of 11 (18%) resection specimens. Among the 19 patients, the 12-month progression-free survival was 58%, and 12-month overall survival was 79%. Common adverse events were alopecia, nausea, vomiting, fatigue, myalgia, peripheral neuropathy, rash, and neutropenia. CONCLUSION Gemcitabine and nab-paclitaxel followed by long-course chemoradiation represents a feasible neoadjuvant treatment strategy for borderline resectable or node-positive pancreatic cancer.
Collapse
Affiliation(s)
- Emerson Y. Chen
- Division of Hematology and Medical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
| | - Adel Kardosh
- Division of Hematology and Medical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
| | - Nima Nabavizadeh
- Department of Radiation MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Bryan Foster
- Department of Diagnostic RadiologyOregon Health & Science UniversityPortlandOregonUSA
| | - Skye C. Mayo
- Division of Surgical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
| | | | - Erin W. Gilbert
- Division of Gastrointestinal and General SurgeryOregon Health & Science UniversityPortlandOregonUSA
| | | | - Aaron Grossberg
- Department of Radiation MedicineOregon Health & Science UniversityPortlandOregonUSA
| | | | | | - Eric C. Anderson
- Division of Hematology and Medical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
| | - Brett C. Sheppard
- Division of Gastrointestinal and General SurgeryOregon Health & Science UniversityPortlandOregonUSA
| | - Charles R. Thomas
- Department of Radiation MedicineOregon Health & Science UniversityPortlandOregonUSA
- Radiation OncologyGeisel School of Medicine at Dartmouth and Dartmouth Cancer CenterNew HampshireLebanonUSA
| | - Charles D. Lopez
- Division of Hematology and Medical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
| | - Gina M. Vaccaro
- Division of Hematology and Medical OncologyOregon Health & Science University, Knight Cancer InstitutePortlandOregonUSA
- Providence Cancer InstitutePortlandOregonUSA
| | | |
Collapse
|
4
|
Piquemal D, Bruno R, Bournet B, Ghiringhelli F, Noguier F, Canivet C, Bertaut A, Pierrat F, Evesque L, Gamez A, Cros J, Rederstorff E, Petit E, Adnet J, Saint A, Drouillard A, Kempf E, Soularue E, Vincent J, Baumgaertner I, Hennequin A, Tournigand C, Lopez Trabada Ataz D, Bengrine L, Lepage C, Manfredi S, Afchain P, Trouilloud I, Gagnaire A, LoConte NK, Bachet JB. Performance of a blood-based RNA signature for gemcitabine-based treatment in metastatic pancreatic adenocarcinoma. J Gastrointest Oncol 2023; 14:997-1007. [PMID: 37201091 PMCID: PMC10186541 DOI: 10.21037/jgo-22-946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer, and chemotherapy is a key treatment for advanced PDAC. Gemcitabine chemotherapy is still an important component of treatment; however, there is no routine biomarker to predict its efficacy. Predictive tests may help clinicians to decide on the best first-line chemotherapy. Methods This study is a confirmatory study of a blood-based RNA signature, called the GemciTest. This test measures the expression levels of nine genes using real-time polymerase chain reaction (PCR) processes. Clinical validation was carried out, through a discovery and a validation phases, on 336 patients (mean 68.7 years; range, 37-88 years) for whom blood was collected from two prospective cohorts and two tumor biobanks. These cohorts included previously untreated advanced PDAC patients who received either a gemcitabine- or fluoropyrimidine-based regimen. Results Gemcitabine-based treated patients with a positive GemciTest (22.9%) had a significantly longer progression-free survival (PFS) {5.3 vs. 2.8 months; hazard ratio (HR) =0.53 [95% confidence interval (CI): 0.31-0.92]; P=0.023} and overall survival (OS) [10.4 vs. 4.8 months; HR =0.49 (95% CI: 0.29-0.85); P=0.0091]. On the contrary, fluoropyrimidine-based treated patients showed no significant difference in PFS and OS using this blood signature. Conclusions The GemciTest demonstrated that a blood-based RNA signature has the potential to aid in personalized therapy for PDAC, leading to better survival rates for patients receiving a gemcitabine-based first-line treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Erwan Petit
- Centre Georges François Leclerc, Dijon, France
| | - Johan Adnet
- Centre Georges François Leclerc, Dijon, France
| | | | | | | | | | | | | | | | | | | | | | - Come Lepage
- CHU Dijon Bourgogne - Hospital François Mitterrand, Dijon, France
| | - Sylvain Manfredi
- CHU Dijon Bourgogne - Hospital François Mitterrand, Dijon, France
| | | | | | - Alice Gagnaire
- CHU Dijon Bourgogne - Hospital François Mitterrand, Dijon, France
| | | | | |
Collapse
|
5
|
de Jong TL, Koopman D, van der Worp CAJ, Stevens H, Vuijk FA, Vahrmeijer AL, Mieog JSD, de Groot JWB, Meijssen MAC, Nieuwenhuijs VB, de Geus-Oei LF, Jager PL, Patijn GA. Added value of digital FDG-PET/CT in disease staging and restaging in patients with resectable or borderline resectable pancreatic cancer. Surg Oncol 2023; 47:101909. [PMID: 36739788 DOI: 10.1016/j.suronc.2023.101909] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/09/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND We studied the added value of digital FDG-PET/CT in disease staging and restaging compared to the standard work-up with contrast enhanced CT (ceCT) and CA19-9 in patients with resectable or borderline resectable pancreatic cancer who received neo-adjuvant therapy. Primary endpoints were tumor response compared to ceCT and CA19.9 as well as the ability to detect distant metastatic disease. METHODS 35 patients were included in this dual-center prospective study. FDG-PET using digital photon counting technology combined with CT scans were acquired before (T1) and after neo-adjuvant therapy (T2). Patients were staged and restaged based on standard protocol with ceCT and CA 19.9, while all PET/CT scans were stored securely and not included in clinical decision making. After the pancreatic resection, an expert team retrospectively assessed the CT tumor diameter, CA19-9, tumor FDG-uptake, and appearance of metastatic disease of all patients for both time points. RESULTS CA19-9 levels, CT tumor diameter, and tumor FDG-uptake on PET significantly decreased from T1 to T2 (p = 0.017, p = 0.001, and p < 0.0001). The change in FDG-uptake values showed a strong positive correlation with the change in CT tumor diameter and change in CA19-9 (R = 0.75 and R = 0.73, respectively). In addition, small-volume liver lesions were detected on digital PET/CT in 5/35 patients (14%), 4 of which were pathology confirmed at laparotomy. Only one of these five cases was detected on baseline staging ceCT (3%). CONCLUSION We found that adding digital PET/CT strengthens restaging after neo-adjuvant therapy based on the observed strong correlation with ceCT tumor diameter and Ca19.9. Also, digital PET/CT was found to detect occult metastatic disease not visualized on ceCT, that would have resulted in altered disease staging and therapeutic strategy in a substantial proportion of patients.
Collapse
Affiliation(s)
- Tonke L de Jong
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | - Daniëlle Koopman
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | | | - Henk Stevens
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | - Floris A Vuijk
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Maarten A C Meijssen
- Department of Gastroenterology and Hepatology, Isala Hospital, Zwolle, the Netherlands
| | | | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Biomedical Photonic Imaging Group, University of Twente, Enschede, the Netherlands
| | - Pieter L Jager
- Department of Nuclear Medicine, Isala Hospital, Zwolle, the Netherlands
| | - Gijs A Patijn
- Department of Surgery, Isala Hospital, Zwolle, the Netherlands.
| |
Collapse
|
6
|
Bruckner HW, Bassali F, Dusowitz E, Gurell D, Book A, De Jager R. Actionable tests and treatments for patients with gastrointestinal cancers and historically short median survival times. PLoS One 2022; 17:e0276492. [PMID: 36322580 PMCID: PMC9629612 DOI: 10.1371/journal.pone.0276492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Patients have difficult unmet needs when standard chemotherapy produces a median survival of less than 1 year or many patients will experience severe toxicities. Blood tests can predict their survival. METHODS Analyses evaluate predictive blood tests to identify patients who often survive 1 and 2 years. A four-test model includes: albumin, absolute neutrophil count, neutrophil-lymphocyte ratio, and lymphocyte-monocyte ratio. Individual tests include: alkaline phosphatase, lymphocytes, white blood count, platelet count, and hemoglobin. Eligible patients have advanced: resistant 3rd line colorectal, and both resistant and new pancreatic and intrahepatic bile duct cancers. Eligibility characteristics include: biopsy-proven, measurable metastatic disease, NCI grade 0-2 blood tests, Karnofsky Score 100-50, and any adult age. Drugs are given at 1/4-1/3 of their standard dosages biweekly: gemcitabine, irinotecan, fluorouracil, leucovorin, and day 2 oxaliplatin every 2 weeks. In case of progression, Docetaxel is added (except colon cancer), with or without Mitomycin C, and next cetuximab (except pancreatic and KRAS BRAF mutation cancers). Bevacizumab is substituted for cetuximab in case of another progression or ineligibility. Consent was written and conforms with Helsinki, IRB, and FDA criteria (FDA #119005). RESULTS Median survival is 14.5 months. Of 205 patients, 60% survive 12, and 37% survive 24 months (95% CI ± 8%). Survival is > 24, 13, and 3.8 months for patients with 0, 1-2, and 3-4 unfavorable tests, respectively. Individual "favorable and unfavorable" tests predict long and short survival. Neither age nor prior therapy discernibly affects survival. Net rates of clinically significant toxicities are less than 5%. CONCLUSION Treatments reproduce predictable, greater than 12 and 24-month chances of survival for the aged and for patients with drug-resistant tumors. Evaluation of blood tests may change practice, expand eligibility, and personalize treatments. Findings support investigation of drug combinations and novel dosages to reverse resistance and improve safety.
Collapse
Affiliation(s)
- Howard W. Bruckner
- MZB Foundation for Cancer Research, New York, NY, United States of America
| | - Fred Bassali
- MZB Foundation for Cancer Research, New York, NY, United States of America
| | - Elisheva Dusowitz
- MZB Foundation for Cancer Research, New York, NY, United States of America
| | - Daniel Gurell
- Department of Diagnostic Radiology, University Diagnostic Imaging, Bronx, New York, United States of America
| | - Abe Book
- MZB Foundation for Cancer Research, New York, NY, United States of America
| | - Robert De Jager
- MZB Foundation for Cancer Research, New York, NY, United States of America
| |
Collapse
|
7
|
Caliez O, Pietrasz D, Ksontini F, Doat S, Simon JM, Vaillant JC, Taly V, Laurent-Puig P, Bachet JB. Circulating tumor DNA: a help to guide therapeutic strategy in patients with borderline and locally advanced pancreatic adenocarcinoma? Dig Liver Dis 2022; 54:1428-1436. [PMID: 35120842 DOI: 10.1016/j.dld.2022.01.126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 12/16/2021] [Accepted: 01/11/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND prognostic biomarkers could be useful to better select patients with borderline resectable (BR) or locally advanced (LA) pancreatic adenocarcinoma (PA) for chemoradiotherapy (CRT) and/or secondary resection. AIMS The main objective of this work was to study characteristics, received treatments and prognostic of patients with BR or LA PA according to their baseline circulating tumor DNA status and, for secondary objective, neutrophil-to-lymphocyte Ratio (NLR). METHODS ctDNA status at baseline was determined using Next Generation Sequencing in a consecutive monocentric cohort of patients with a BR or LA PA. RESULTS 69 patients were included, 31 with BR PA and 38 with LA PA. 14 (20.3%) patients had baseline positive ctDNA. Five (7.8%) patients had NLR> 5. Patients with positive ctDNA had 3.7 months shorter progression free survival (p = 0.006). Patients with positive ctDNA had earlier progression after the beginning of CRT (4.4 vs 7.1 months; p = 0.068) and shorter relapse free survival after secondary resection (9.2 vs 22.9 months; p = 0.016). CONCLUSIONS positive ctDNA at baseline was associated with a worse prognosis in patients with BR or LA PA. These data are exploratory and must be confirmed in further prospective trials.
Collapse
Affiliation(s)
- Olivier Caliez
- Department of Gastroenterology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Sorbonne Université, UPMC, Paris 6, France
| | - Daniel Pietrasz
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Department of Digestive Surgery, Hôpital Paul Brousse, Villejuif, France
| | - Feryel Ksontini
- Department of Oncology, Institute Salah-Azaïz, Tunis, Tunisia
| | - Solène Doat
- Department of Gastroenterology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean-Marc Simon
- Department of Radiation Oncology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean-Christophe Vaillant
- Department of Digestive Surgery, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Valerie Taly
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France
| | - Pierre Laurent-Puig
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France
| | - Jean-Baptiste Bachet
- Department of Gastroenterology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Sorbonne Université, UPMC, Paris 6, France.
| |
Collapse
|
8
|
Abdelrahman AM, Goenka AH, Alva-Ruiz R, Yonkus JA, Leiting JL, Graham RP, Merrell KW, Thiels CA, Hallemeier CL, Warner SG, Haddock MG, Grotz TE, Tran NH, Smoot RL, Ma WW, Cleary SP, McWilliams RR, Nagorney DM, Halfdanarson TR, Kendrick ML, Truty MJ. FDG-PET Predicts Neoadjuvant Therapy Response and Survival in Borderline Resectable/Locally Advanced Pancreatic Adenocarcinoma. J Natl Compr Canc Netw 2022; 20:1023-1032.e3. [PMID: 36075389 DOI: 10.6004/jnccn.2022.7041] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/03/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Neoadjuvant therapy (NAT) is used in borderline resectable/locally advanced (BR/LA) pancreatic ductal adenocarcinoma (PDAC). Anatomic imaging (CT/MRI) poorly predicts response, and biochemical (CA 19-9) markers are not useful (nonsecretors/nonelevated) in many patients. Pathologic response highly predicts survival post-NAT, but is only known postoperatively. Because metabolic imaging (FDG-PET) reveals primary tumor viability, this study aimed to evaluate our experience with preoperative FDG-PET in patients with BR/LA PDAC in predicting NAT response and survival. METHODS We reviewed all patients with resected BR/LA PDAC who underwent NAT with FDG-PET within 60 days of resection. Pre- and post-NAT metabolic (FDG-PET) and biochemical (CA 19-9) responses were dichotomized in addition to pathologic responses. We compared post-NAT metabolic and biochemical responses as preoperative predictors of pathologic responses and recurrence-free survival (RFS) and overall survival (OS). RESULTS We identified 202 eligible patients. Post-NAT, 58% of patients had optimization of CA 19-9 levels. Major metabolic and pathologic responses were present in 51% and 38% of patients, respectively. Median RFS and OS times were 21 and 48.7 months, respectively. Metabolic response was superior to biochemical response in predicting pathologic response (area under the curve, 0.86 vs 0.75; P<.001). Metabolic response was the only univariate preoperative predictor of OS (odds ratio, 0.25; 95% CI, 0.13-0.40), and was highly correlated (P=.001) with pathologic response as opposed to biochemical response alone. After multivariate adjustment, metabolic response was the single largest independent preoperative predictor (P<.001) for pathologic response (odds ratio, 43.2; 95% CI, 16.9-153.2), RFS (hazard ratio, 0.37; 95% CI, 0.2-0.6), and OS (hazard ratio, 0.21; 95% CI, 0.1-0.4). CONCLUSIONS Among patients with post-NAT resected BR/LA PDAC, FDG-PET highly predicts pathologic response and survival, superior to biochemical responses alone. Given the poor ability of anatomic imaging or biochemical markers to assess NAT responses in these patients, FDG-PET is a preoperative metric of NAT efficacy, thereby allowing potential therapeutic alterations and surgical treatment decisions. We suggest that FDG-PET should be an adjunct and recommended modality during the NAT phase of care for these patients.
Collapse
Affiliation(s)
| | - Ajit H Goenka
- Division of Nuclear Medicine Radiology, Department of Radiology
| | - Roberto Alva-Ruiz
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | - Jennifer A Yonkus
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | | | - Rondell P Graham
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology
| | | | | | | | - Susanne G Warner
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | | | - Travis E Grotz
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | - Nguyen H Tran
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Rory L Smoot
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | - Wen Wee Ma
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Sean P Cleary
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | - Robert R McWilliams
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - David M Nagorney
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| | | | | | - Mark J Truty
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery
| |
Collapse
|
9
|
Wang Y, Wang Z, Li K, Xiang W, Chen B, Jin L, Hao K. lncRNAs Functioned as ceRNA to Sponge miR-15a-5p Affects the Prognosis of Pancreatic Adenocarcinoma and Correlates With Tumor Immune Infiltration. Front Genet 2022; 13:874667. [PMID: 35899199 PMCID: PMC9312832 DOI: 10.3389/fgene.2022.874667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is one of the most common malignant tumors with poor prognosis worldwide. Mounting evidence suggests that the expression of lncRNAs and the infiltration of immune cells have prognostic value for patients with PAAD. We used Gene Expression Omnibus (GEO) database and identified six genes (COL1A2, ITGA2, ITGB6, LAMA3, LAMB3, and LAMC2) that could affect the survival rate of pancreatic adenocarcinoma patients. Based on a series of in silico analyses for reverse prediction of target genes associated with the prognosis of PAAD, a ceRNA network of mRNA (COL1A2, ITGA2, LAMA3, LAMB3, and LAMC2)–microRNA (miR-15a-5p)–long non-coding RNA (LINC00511, LINC01578, PVT1, and TNFRSF14-AS1) was constructed. We used the algorithm “CIBERSORT” to assess the proportion of immune cells and found three overall survival (OS)–associated immune cells (monocytes, M1 macrophages, and resting mast cell). Moreover, the OS-associated gene level was significantly positively associated with immune checkpoint expression and biomarkers of immune cells. In summary, our results clarified that ncRNA-mediated upregulation of OS-associated genes and tumor-infiltration immune cells (monocytes, M1 macrophages M1, and resting mast cell resting) correlated with poor prognosis in PAAD.
Collapse
Affiliation(s)
- Yu Wang
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhen Wang
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - KaiQiang Li
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - WeiLing Xiang
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - BinYu Chen
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - LiQin Jin
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
- Department of Scientific Research, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
- *Correspondence: LiQin Jin, ; Ke Hao,
| | - Ke Hao
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
- *Correspondence: LiQin Jin, ; Ke Hao,
| |
Collapse
|
10
|
Hsa_circ_0000994 Inhibits Pancreatic Cancer Progression by Clearing Immune-Related miR-27a and miR-27b. JOURNAL OF ONCOLOGY 2022; 2022:7274794. [PMID: 35669238 PMCID: PMC9166970 DOI: 10.1155/2022/7274794] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/23/2022]
Abstract
Pancreatic cancer (PC) is a common cause of cancer death. Although more and more evidences suggest that circular RNAs (circRNAs) are associated with the development of cancer, the biological function of circRNAs in PC has not been fully explored. Based on previous studies, Hsa_circ_0000994 was screened out, and its clinical significance, functional role, and mechanism in PC are poorly studied. In various cell lines, 50 PC tissues, and an equal number of normal tissues, RT-qPCR was used to identify expression level of Hsa_circ_0000994. The impact of Hsa_circ_0000994 on metastasis, cell proliferation, and apoptosis was detected using functional loss and functional gain tests. An animal study was also conducted. Underlying mechanisms of Hsa_circ_0000994 were revealed by luciferase reporter gene detection. Hsa_circ_0000994 was lowly expressed in PC tissues as well as various PC cell lines, and this low expression was closely related to cancer. In terms of functional testing, Hsa_circ_0000994 suppressed core ability of PC cells, including proliferation, migration, and invasion ability. Xenotransplantation studies further confirmed the effect of Hsa_circ_0000994 in promoting cell growth. Mechanically, Hsa_circ_0000994 inhibited miR-27a and miR-27b. Hsa_circ_0000994 inhibited the cancer cells through the effect on miR-27a and miR-27b. In summary, a circRNA with tumor suppressor effects on PC has been elucidated.
Collapse
|
11
|
Prognostic Implications of Intratumoral and Peritumoral Infiltrating Lymphocytes in Pancreatic Ductal Adenocarcinoma. Curr Oncol 2021; 28:4367-4376. [PMID: 34898543 PMCID: PMC8628731 DOI: 10.3390/curroncol28060371] [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: 10/12/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 11/17/2022] Open
Abstract
This study aimed to elucidate the prognostic implications of intratumoral and peritumoral infiltrating T-lymphocytes in pancreatic ductal adenocarcinoma (PDAC) through a meta-analysis. A total of 18 eligible studies and 2453 PDAC patients were included in the present study. Intratumoral and peritumoral infiltrating lymphocytes were evaluated using various markers, such as CD3, CD4, CD8, FOXP3, and immune cell score. The correlations between these parameters and overall and disease-free survival were investigated and used in the meta-analysis. High intratumoral infiltration of CD3-, CD4-, and CD8-expressing lymphocytes was significantly correlated with better overall survival (hazard ratio (HR) 0.747, 95% confidence interval (CI) 0.620-0.900, HR 0.755, 95% CI 0.632-0.902, and HR 0.754, 95% CI 0.611-0.930, respectively). However, there was no significant correlation between PDAC prognosis and intratumoral FOXP3 or immune cell score (HR 1.358, 95% CI 1.115-1.655 and HR 0.776, 95% CI 0.566-1.065, respectively). Moreover, there was no significant correlation between the prognosis and peritumoral infiltrating T-lymphocytes. In evaluations of disease-free survival, only high intratumoral CD4 infiltration was correlated with a better prognosis (HR 0.525, 95% CI 0.341-0.810). Our results showed that high intratumoral infiltrating lymphocytes were significantly correlated with a better PDAC prognosis. However, among the tumor-infiltrating lymphocytes, CD3, CD4, and CD8 had prognostic implications, but not FOXP3 and immune cell score.
Collapse
|
12
|
Williet N, Petrillo A, Roth G, Ghidini M, Petrova M, Forestier J, Lopez A, Thoor A, Weislinger L, De Vita F, Taieb J, Phelip JM. Gemcitabine/Nab-Paclitaxel versus FOLFIRINOX in Locally Advanced Pancreatic Cancer: A European Multicenter Study. Cancers (Basel) 2021; 13:cancers13112797. [PMID: 34199796 PMCID: PMC8200096 DOI: 10.3390/cancers13112797] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Gemcitabine/nab-paclitaxel (GN) and FOLFIRINOX (FFX) are two standard first-line therapies for metastatic pancreatic cancer (PC) but have rarely been compared, especially in patients with locally advanced PC (LAPC). By carefully selecting patients, it is likely these two regimens lead to similar survival outcomes. Through a multicenter European study, biases regarding practice habits are reduced. Hence, we observed no difference between GN and FFX as first-line treatments in patients with LAPC in terms of either survival, tumor response or tumor resection rate. Further trials are needed to confirm these data. Abstract Background: Gemcitabine/nab-paclitaxel (GN) and FOLFIRINOX (FFX) are two standard first-line therapies for metastatic pancreatic cancer (PC) but have rarely been compared, especially in patients with locally advanced PC (LAPC). Methods: This is a retrospective European multicenter study including patients with LAPC treated with either GN or FFX as the first-line therapy between 2010 and 2019. Coprimary objectives were progression-free survival (PFS) and overall survival (OS), both estimated using the Kaplan–Meier method. Results: A total of 147 patients (GN: n = 60; FFX: n = 87) were included. Tumor resection rates were similar between the two groups (16.7% vs. 16.1%; p = 1), with similar R0 resection rates (88.9%). Median PFS rates were not statistically different: 9 months (95% CI: 8–13.5) vs. 12.1 months (95% CI: 10.1–14.6; p = 0.8), respectively. Median OS rates were 15.7 months (95% CI: 12.6–20.2) and 16.7 months (95% CI: 14.8–20.4; p = 0.7), respectively. Abdominal pain at the baseline (HR = 2.03, p = 0.03), tumors located in the tail of the pancreas (HR = 4.35, p = 0.01), CA19-9 > 200 UI/L (HR = 2.03, p = 0.004) and tumor resection (HR = 0.37, p = 0.007) were independent prognostic factors for PFS, similarly to OS. CA19-9 ≤ 200 UI/L (OR = 2.6, p = 0.047) was predictive of the tumor response. Consolidation chemoradiotherapy, more often used in the FFX group (11.7% vs. 50.6%; p < 0.001), was not predictive. Conclusion: This retrospective study did not show any difference between GN and FFX as the first-line treatment in patients with LAPC.
Collapse
Affiliation(s)
- Nicolas Williet
- Department of Hepatogastroenterology, University Hospital of Saint-Etienne, 42000 Saint-Etienne, France;
- Correspondence:
| | - Angelica Petrillo
- Department of Precision Medecine, University of Study of Campania «L. Vanvitelli», 81100 Naples, Italy; (A.P.); (F.D.V.)
| | - Gaël Roth
- Hepato-Gastroenterology Department, University Hospital of Grenoble, 38043 Grenoble, France; (G.R.); (A.T.)
| | - Michele Ghidini
- Department of Medical Oncology, Cancer Center, Hospital of Cremona, 26100 Cremona, Italy;
| | - Mila Petrova
- Department of Medical Oncology, MHAT Nadezhda, 1220 Sofia, Bulgaria;
| | - Julien Forestier
- Department of Medical Oncology, Hôpital Edouard Herriot, 69622 Lyon, France;
| | - Anthony Lopez
- Hepato-Gastroenterology Department, University Hospital of Nancy, 54500 Vandoeuvre-lès-Nancy, France; (A.L.); (L.W.)
| | - Audrey Thoor
- Hepato-Gastroenterology Department, University Hospital of Grenoble, 38043 Grenoble, France; (G.R.); (A.T.)
| | - Lucie Weislinger
- Hepato-Gastroenterology Department, University Hospital of Nancy, 54500 Vandoeuvre-lès-Nancy, France; (A.L.); (L.W.)
| | - Ferdinando De Vita
- Department of Precision Medecine, University of Study of Campania «L. Vanvitelli», 81100 Naples, Italy; (A.P.); (F.D.V.)
| | - Julien Taieb
- Department of Gastroenterology and Gastro-Intestinal Oncology, Hôpital Européen Georges-Pompidou, APHP, Paris Descartes University, Sorbonne Paris Cité, 75004 Paris, France;
| | - Jean Marc Phelip
- Department of Hepatogastroenterology, University Hospital of Saint-Etienne, 42000 Saint-Etienne, France;
| |
Collapse
|
13
|
Wang C, Wang J, Cui W, Liu Y, Zhou H, Wang Y, Chen X, Chen X, Wang Z. Serum Exosomal miRNA-1226 as Potential Biomarker of Pancreatic Ductal Adenocarcinoma. Onco Targets Ther 2021; 14:1441-1451. [PMID: 33664577 PMCID: PMC7924134 DOI: 10.2147/ott.s296816] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related mortality and it is urgent to find biomarkers for early detection of PDAC. Exosomal miRNAs are useful biomarkers for cancer detection. The aims of this study were to investigate the potential role of serum exosomal miRNA in detection of PDAC and to analyze the correlation between the levels of exosome miRNA and the tumor biological behaviors. Materials and Methods Thirteen serum samples were collected from five patients with PDACs, three healthy individuals (HIs) and five benign pancreatic lesions (BP) for a high throughput profiling analysis to identify an altered miRNA expression patterns in PDAC. Candidate exosomal miRNAs were filtered based on a second independent cohort that included 17 PDACs and 12 benign pancreatic lesions by quantitative real-time polymerase chain reaction (qRT-PCR). Four miRNAs were selected for miRNA validation as PDAC biomarkers in a subsequent set of samples. The association between candidate exosomal miRNA and tumor behavior (tumor invasion or metastases) was evaluated in 17 PDACs. In vitro studies were performed to evaluate the role of candidate exosomal miRNA on cell viability, apoptosis and cell migration in two PDAC cell lines. Results The expression of 11 miRNAs showed same trend between PDAC and BP, and between PDAC and HIs. Six of them were upregulated (miR-203b-5p, miR-342-5p, miR-337-5p, miR-149-5p, miR-877-5p, miR-203a-3p), and five were downregulated (miR-1226-3p, miR-3182, miR-625-3p, miR-624-5p, miR-664a-5p). miR-1226-3p was selected as the candidate exosomal biomarker for the PDAC detection. The expression of serum exosomal miRNA-1226-3p was downregulated in PDACs compared to the BPs (p = 0.025). miR-1226-3p had acceptable performance in predicting [area under the curve (AUC) = 0.74] PDAC. Exosomal miRNA-1226-3p level in PDAC with invasion or metastases was lower than that without invasion or metastases (p = 0.028). Transfection of miRNA-1226-3p significantly inhibited the proliferation of PANC-1 and BXP-3 cells, stimulated cell apoptosis and inhibited cell migration. Conclusion Serum exosomal miRNA-1226-3p is a potential biomarker in diagnosing and predicting the tumor invasion or metastases of PDAC.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China.,Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210029, People's Republic of China
| | - Jianhua Wang
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China
| | - Wenjing Cui
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China
| | - Yongkang Liu
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China
| | - Hao Zhou
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China
| | - Yajie Wang
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China
| | - Xin Chen
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China
| | - Xiao Chen
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China
| | - Zhongqiu Wang
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China
| |
Collapse
|
14
|
Pallerla S, Abdul AURM, Comeau J, Jois S. Cancer Vaccines, Treatment of the Future: With Emphasis on HER2-Positive Breast Cancer. Int J Mol Sci 2021; 22:E779. [PMID: 33466691 PMCID: PMC7828795 DOI: 10.3390/ijms22020779] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is one of the leading causes of death in women. With improvements in early-stage diagnosis and targeted therapies, there has been an improvement in the overall survival rate in breast cancer over the past decade. Despite the development of targeted therapies, tyrosine kinase inhibitors, as well as monoclonal antibodies and their toxin conjugates, all metastatic tumors develop resistance, and nearly one-third of HER2+ breast cancer patients develop resistance to all these therapies. Although antibody therapy has shown promising results in breast cancer patients, passive immunotherapy approaches have limitations and need continuous administration over a long period. Vaccine therapy introduces antigens that act on cancer cells causing prolonged activation of the immune system. In particular, cancer relapse could be avoided due to the presence of a longer period of immunological memory with an effective vaccine that can protect against various tumor antigens. Cancer vaccines are broadly classified as preventive and therapeutic. Preventive vaccines are used to ward off any future infections and therapeutic vaccines are used to treat a person with active disease. In this article, we provided details about the tumor environment, different types of vaccines, their advantages and disadvantages, and the current status of various vaccine candidates with a focus on vaccines for breast cancer. Current data indicate that therapeutic vaccines themselves have limitations in terms of efficacy and are used in combination with other chemotherapeutic or targeting agents. The majority of breast cancer vaccines are undergoing clinical trials and the next decade will see the fruitfulness of breast cancer vaccine therapy.
Collapse
Affiliation(s)
- Sandeep Pallerla
- School of Pharmaceutical and Toxicological Sciences and School of Clinical Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA; (S.P.); (J.C.)
| | | | - Jill Comeau
- School of Pharmaceutical and Toxicological Sciences and School of Clinical Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA; (S.P.); (J.C.)
| | - Seetharama Jois
- School of Pharmaceutical and Toxicological Sciences and School of Clinical Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA; (S.P.); (J.C.)
| |
Collapse
|
15
|
|
16
|
Han B, Kim BJ, Kim HS, Choi DR, Shim BY, Lee KH, Kim JW, Kim JH, Song H, Kim JH, Park CK, Lee JW, Kim MJ, Zang DY. A phase II study of gemcitabine, erlotinib and S-1 in patients with advanced pancreatic cancer. J Cancer 2021; 12:912-917. [PMID: 33403047 PMCID: PMC7778528 DOI: 10.7150/jca.50514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/01/2020] [Indexed: 12/26/2022] Open
Abstract
Background: We evaluated the efficacy and safety of gemcitabine in combination with erlotinib and S-1 for the treatment of advanced pancreatic cancer. Methods: Chemotherapy-naïve patients with pathologically-proven locally advanced, recurrent, or metastatic pancreatic adenocarcinoma were assessed for eligibility. Gemcitabine was administered at 1,000 mg/m2 intravenously on days 1 and 8, erlotinib was administered at 100 mg/day on days 1-21, and S-1 was administered at 60 mg/m2 on days 1-14 every 21 days and continued to a maximum of 8 cycles of treatment. Dose escalation of S-1 to 80 mg/m2 was permitted from the second cycle for pre-defined tolerable patients. Results: Thirty-seven patients (median age 61.5 years) were enrolled. A total of 140 cycles of chemotherapy were administered (median of 3.8; range 1-8 cycles). Toxicities were evaluated in 36 patients, and the responses were evaluated in 32 patients. Major grade 3/4 toxicities included neutropenia (25%), febrile neutropenia (2.8%), fatigue (22.2%), infection (8.3%), vomiting (5.6%), and mucositis (5.6%). The confirmed overall response rate was 12.5% [95% confidence interval (CI), 5.1-28.9%] and the disease control rate was 71.9% (95% CI, 56.8-86.3%). The median progression-free survival and overall survival were 3.7 months (95% CI, 2.8-4.6 months) and 6.7 months (95% CI, 3.4-9.9 months), respectively. Conclusion: The combination of gemcitabine, erlotinib, and S-1 provided an acceptable toxicity profile and modest clinical benefits in patients with advanced pancreatic cancer.
Collapse
Affiliation(s)
- Boram Han
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeongigi-do, Korea
| | - Bum Jun Kim
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeongigi-do, Korea
| | - Hyeong Su Kim
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeongigi-do, Korea
| | - Dae Ro Choi
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeongigi-do, Korea
| | - Byoung Yong Shim
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Kyung Hee Lee
- Department of Internal Medicine, Yeungnam University Medical Center, Yeungnam University College of Medicine, Daegu, Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Gyeonggi-do, Korea
| | - Jung Han Kim
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeongigi-do, Korea
| | - Hunho Song
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeongigi-do, Korea
| | - Jong Hyeok Kim
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeongigi-do, Korea
| | - Choong Kee Park
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeongigi-do, Korea
| | - Jung Woo Lee
- Department of General Surgery, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeongigi-do, Korea
| | - Min-Jeong Kim
- Departement of Radiology, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeongigi-do, Korea
| | - Dae Young Zang
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeongigi-do, Korea
| |
Collapse
|
17
|
Jiang S, Li DL, Chen J, Zheng X, Wu PP, Li C, Xu XT, Zhang K. Synergistic Anticancer Effect of Gemcitabine Combined With Impressic Acid or Acankoreanogein in Panc-1 Cells by Inhibiting NF-κB and Stat 3 Activation. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20974239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Natural products have presented potentiality to improve the outcomes of cancer therapies. Impressic acid (E12-1) and acankoreanogein (E13-1), important activity compounds in Acanthopanax trifoliatus (L.) Merr., show widely biological activities. In this study, we isolated E12-1 and E13-1 from Acanthopanax trifoliatus (L.) Merr., and investigated their improvement effect in gemcitabine (GEM) treatment in Panc-1 cells. The results showed that GEM in combination with E12-1 or E13-1 showed stronger inhibition on the growth and induction of apoptosis in Panc-1 cells compared to GEM, E12-1, or E13-1 alone. GEM in combination with E12-1or E13-1 also strongly inhibited cell migration. Mechanistic investigation showed that GEM in combination with E12-1or E13-1 effectively inhibited the activition of nuclear factor kappa-light-chain-enhancer of activated B cells and Stat 3. Overall, GEM in combination with E12-1 or E13-1 might be an effective strategy for the prevention of prostate cancer.
Collapse
Affiliation(s)
- Sen Jiang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- Guangzhou Baiyunshan Chenliji Pharmaceutical Co., Ltd., China
- Laboratory of Natural Medicinal Chemistry & Green Chemistry, Guangdong University of Technology, China
| | - Dong-Li Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Jie Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Xi Zheng
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Pan-Pan Wu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Chen Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Xue-Tao Xu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Kun Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| |
Collapse
|
18
|
Iovanna J. Implementing biological markers as a tool to guide clinical care of patients with pancreatic cancer. Transl Oncol 2020; 14:100965. [PMID: 33248412 PMCID: PMC7704461 DOI: 10.1016/j.tranon.2020.100965] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
A major obstacle for the effective treatment of PDAC is its molecular heterogeneity. Stratification of PDAC using markers highly specific, reproducible, sensitive, easily measurable and inexpensive is necessary. At the early stages, clinician’s priority lies in rapid diagnosis, so that the patient receives surgery without delay. At advanced disease stages, priority is to determine the tumor subtype and select a suitable effective treatment.
A major obstacle for the effective treatment of pancreatic ductal adenocarcinoma (PDAC) is its molecular heterogeneity, reflected by the diverse clinical outcomes and responses to therapies that occur. The tumors of patients with PDAC must therefore be closely examined and classified before treatment initiation in order to predict the natural evolution of the disease and the response to therapy. To stratify patients, it is absolutely necessary to identify biological markers that are highly specific and reproducible, and easily measurable by inexpensive sensitive techniques. Several promising strategies to find biomarkers are already available or under development, such as the use of liquid biopsies to detect circulating tumor cells, circulating free DNA, methylated DNA, circulating RNA, and exosomes and extracellular vesicles, as well as immunological markers and molecular markers. Such biomarkers are capable of classifying patients with PDAC and predicting their therapeutic sensitivity. Interestingly, developing chemograms using primary cell lines or organoids and analyzing the resulting high-throughput data via artificial intelligence would be highly beneficial to patients. How can exploiting these biomarkers benefit patients with resectable, borderline resectable, locally advanced, and metastatic PDAC? In fact, the utility of these biomarkers depends on the patient's clinical situation. At the early stages of the disease, the clinician's priority lies in rapid diagnosis, so that the patient receives surgery without delay; at advanced disease stages, where therapeutic possibilities are severely limited, the priority is to determine the PDAC tumor subtype so as to estimate the clinical outcome and select a suitable effective treatment.
Collapse
Affiliation(s)
- Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, France.
| |
Collapse
|
19
|
Rhee H, Park MS. The Role of Imaging in Current Treatment Strategies for Pancreatic Adenocarcinoma. Korean J Radiol 2020; 22:23-40. [PMID: 32901458 PMCID: PMC7772381 DOI: 10.3348/kjr.2019.0862] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/30/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
In pancreatic cancer, imaging plays an essential role in surveillance, diagnosis, resectability evaluation, and treatment response evaluation. Pancreatic cancer surveillance in high-risk individuals has been attempted using endoscopic ultrasound (EUS) or magnetic resonance imaging (MRI). Imaging diagnosis and resectability evaluation are the most important factors influencing treatment decisions, where computed tomography (CT) is the preferred modality. EUS, MRI, and positron emission tomography play a complementary role to CT. Treatment response evaluation is of increasing clinical importance, especially in patients undergoing neoadjuvant therapy. This review aimed to comprehensively review the role of imaging in relation to the current treatment strategy for pancreatic cancer, including surveillance, diagnosis, evaluation of resectability and treatment response, and prediction of prognosis.
Collapse
Affiliation(s)
- Hyungjin Rhee
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Suk Park
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
20
|
Piehler S, Wucherpfennig L, Tansi FL, Berndt A, Quaas R, Teichgraeber U, Hilger I. Hyperthermia affects collagen fiber architecture and induces apoptosis in pancreatic and fibroblast tumor hetero-spheroids in vitro. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2020; 28:102183. [PMID: 32222478 DOI: 10.1016/j.nano.2020.102183] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/04/2020] [Accepted: 03/14/2020] [Indexed: 12/16/2022]
Abstract
Desmoplasia, an aberrant production of extracellular matrix (ECM), is considered as one predictive marker of malignancy of pancreatic cancer. In this paper, we study the effect of mild hyperthermia on fibrillary collagen architecture in murine Achilles tendons and in a pancreatic cancer model, in vitro, i.e. 3D hetero-type tumor spheroids, consisting of pancreatic cancer (Panc-1) cells and fibroblasts (WI-38), producing collagen fibers. We clearly demonstrate that i) mild hyperthermia (40 °C, 42 °C) damages the collagen architecture in murine Achilles tendons. ii) Mild extrinsic (hot air) and iron oxide nanoparticle based magnetic hyperthermia reduce the level of collagen fiber architecture in the generated hetero-type tumor spheroids. iii) Mild magnetic hyperthermia reduces cell vitality mainly through apoptotic and necrotic processes in the generated tumor spheroids. In conclusion, hetero-type 3D tumor spheroids are suitable for studying the effect of hyperthermia on collagen fibers, in vitro.
Collapse
Affiliation(s)
- Susann Piehler
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Jena, Germany
| | - Lena Wucherpfennig
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Jena, Germany
| | - Felista L Tansi
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Jena, Germany
| | - Alexander Berndt
- Section Pathology, Institute of Legal Medicine, Jena University Hospital, Jena, Germany
| | | | - Ulf Teichgraeber
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Jena, Germany
| | - Ingrid Hilger
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Jena, Germany.
| |
Collapse
|
21
|
Mas L, Schwarz L, Bachet JB. Adjuvant chemotherapy in pancreatic cancer: state of the art and future perspectives. Curr Opin Oncol 2020; 32:356-363. [PMID: 32541325 DOI: 10.1097/cco.0000000000000639] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW The modalities of management of resectable pancreatic ductal adenocarcinoma (PDAC) have evolved in recent years with new practice guidelines on adjuvant chemotherapy and results of randomized phase III trials. The aim of this review is to describe the state of the art in this setting and to highlight future possible perspectives. RECENT FINDINGS Resectable PDAC is the tumor without vascular contact or a limited venous contact without vein irregularity. Several pathologic and biologic robust prognostic factors such as an R0 resection defined by a margin at least 1 mm have been validated. In phase III trials, the doublet gemcitabine-capecitabine provided a statistically significant, albeit modest overall survival benefit, but failed to show an improvement in relapse-free survival. Similarly, gemcitabine plus nab-paclitaxel did not increase disease-free survival. Modified FOLFIRINOX led to improved disease-free survival, overall survival, and metastasis-free survival, with acceptable toxicity. In the future, prognostic and/or predictive biomarkers could lead the optimization of therapeutic strategies and neoadjuvant treatment could become a standard of care in PDAC. SUMMARY After curative intent resection, modified FOLFIRINOX is the standard of care in adjuvant in fit patients with PDAC. Others regimens (monotherapy or gemcitabine-based) are an option in unfit patients.
Collapse
Affiliation(s)
- Léo Mas
- Department of Hepato-gastroenterology, Groupe Hospitalier Pitié Salpêtrière, Paris
| | - Lilian Schwarz
- Department of Digestive Surgery, Rouen University Hospital
- Department of Genomic and Personalized Medicine in Cancer and Neurological Disorders, Normandie University, UNIROUEN, UMR 1245 INSERM, Rouen University Hospital, Rouen
| | - Jean-Baptiste Bachet
- Department of Hepato-gastroenterology, Groupe Hospitalier Pitié Salpêtrière, Paris
- Sorbonne University, UPMC University, Paris, France
| |
Collapse
|
22
|
Liu YC, Yeh CT, Lin KH. Cancer Stem Cell Functions in Hepatocellular Carcinoma and Comprehensive Therapeutic Strategies. Cells 2020; 9:cells9061331. [PMID: 32466488 PMCID: PMC7349579 DOI: 10.3390/cells9061331] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a significant cause of cancer-related mortality owing to resistance to traditional treatments and tumor recurrence after therapy, which leads to poor therapeutic outcomes. Cancer stem cells (CSC) are a small subset of tumor cells with the capability to influence self-renewal, differentiation, and tumorigenesis. A number of surface markers for liver cancer stem cell (LCSC) subpopulations (EpCAM, CD133, CD44, CD13, CD90, OV-6, CD47, and side populations) in HCC have been identified. LCSCs play critical roles in regulating HCC stemness, self-renewal, tumorigenicity, metastasis, recurrence, and therapeutic resistance via genetic mutations, epigenetic disruption, signaling pathway dysregulation, or alterations microenvironment. Accumulating studies have shown that biomarkers for LCSCs contribute to diagnosis and prognosis prediction of HCC, supporting their utility in clinical management and development of therapeutic strategies. Preclinical and clinical analyses of therapeutic approaches for HCC using small molecule inhibitors, oncolytic measles viruses, and anti-surface marker antibodies have demonstrated selective, efficient, and safe targeting of LCSC populations. The current review focuses on recent reports on the influence of LCSCs on HCC stemness, tumorigenesis, and multiple drug resistance (MDR), along with LCSC-targeted therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Yu-Chin Liu
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan;
- Department of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
| | - Kwang-Huei Lin
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan;
- Department of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Correspondence: ; Tel./Fax: +886-3-211-8263
| |
Collapse
|
23
|
Kong Y, Li Y, Luo Y, Zhu J, Zheng H, Gao B, Guo X, Li Z, Chen R, Chen C. circNFIB1 inhibits lymphangiogenesis and lymphatic metastasis via the miR-486-5p/PIK3R1/VEGF-C axis in pancreatic cancer. Mol Cancer 2020; 19:82. [PMID: 32366257 PMCID: PMC7197141 DOI: 10.1186/s12943-020-01205-6] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/23/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Patients with lymph node (LN)-positive pancreatic ductal adenocarcinoma (PDAC) have extremely poor survival rates. Circular RNAs (circRNAs), a newly discovered type of endogenous noncoding RNAs, have been proposed to mediate the progression of diverse types of tumors. However, the role and underlying regulatory mechanisms of circRNAs in the LN metastasis of PDAC remain unknown. METHODS Next-generation sequencing was used to identify differentially expressed circRNAs between PDAC and normal adjacent tissues. In vitro and in vivo experiments were conducted to evaluate the functional role of circNFIB1. RNA pulldown and luciferase assays were performed to examine the binding of circNFIB1 and miR-486-5p. RESULTS In the present study, we identified that a novel circRNA (circNFIB1, hsa_circ_0086375) was downregulated in PDAC and negatively associated with LN metastasis in PDAC patients. Functionally, circNFIB1 knockdown promoted lymphangiogenesis and LN metastasis of PDAC both in vitro and in vivo. Mechanistically, circNFIB1 functioned as a sponge of miR-486-5p, and partially reversed the effect of miR-486-5p. Moreover, circNFIB1 attenuated the oncogenic effect of miR-486-5p and consequently upregulated PIK3R1 expression, which further downregulated VEGF-C expression through inhibition of the PI3K/Akt pathway, and ultimately suppressed lymphangiogenesis and LN metastasis in PDAC. CONCLUSIONS Our findings provide novel insight into the underlying mechanism of circRNA-mediated LN metastasis of PDAC and suggest that circNFIB1 may serve as a potential therapeutic target for LN metastasis in PDAC.
Collapse
Affiliation(s)
- Yao Kong
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, 510120, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Yuting Li
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Yuming Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, 510120, People's Republic of China.,Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Jiang Zhu
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Hanhao Zheng
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Bowen Gao
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Xiaofeng Guo
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, 510120, People's Republic of China
| | - Zhihua Li
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, 107th Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong, 510120, People's Republic of China.
| | - Rufu Chen
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China.
| | - Changhao Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, 510120, People's Republic of China. .,Department of Urology, Sun Yat-sen Memorial Hospital, 107 Yanjiangxi Road, Yuexiu District, Guangzhou, Guangdong, 510120, People's Republic of China.
| |
Collapse
|
24
|
Sinn M, Sinn BV, Treue D, Keilholz U, Damm F, Schmuck R, Lohneis P, Klauschen F, Striefler JK, Bahra M, Bläker H, Bischoff S, Pelzer U, Oettle H, Riess H, Budczies J, Denkert C. TP53 Mutations Predict Sensitivity to Adjuvant Gemcitabine in Patients with Pancreatic Ductal Adenocarcinoma: Next-Generation Sequencing Results from the CONKO-001 Trial. Clin Cancer Res 2020; 26:3732-3739. [PMID: 32234756 DOI: 10.1158/1078-0432.ccr-19-3034] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/14/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE We performed next-generation sequencing (NGS) in the CONKO-001 phase III trial to identify clinically relevant prognostic and predictive mutations and conducted a functional validation in The Cancer Genome Atlas (TCGA) sequencing data. EXPERIMENTAL DESIGN Patients of the CONKO-001 trial received curatively intended surgery for pancreatic adenocarcinoma (PDAC) followed by adjuvant chemotherapy with gemcitabine (Gem) or observation only (Obs). Tissue samples of 101 patients were evaluated by NGS of 37 genes. Cox proportional hazard models were applied for survival analysis. In addition, functional genomic analyses were performed in an NGS and RNA-sequencing dataset of 146 pancreatic tumors from TCGA. RESULTS The most common mutations in the CONKO cohort were KRAS (75%), TP53 (60%), SMAD4 (10%), CDKNA2 (9%), as well as SWI/SNF (12%) complex alterations. In untreated patients, TP53 mutations were a negative prognostic factor for disease-free survival (DFS; HR mut vs. WT 2.434, P = 0.005). With respect to gemcitabine treatment, TP53 mutations were a positive predictive factor for gemcitabine efficacy [TP53mut: HR for DFS Gem vs. Obs, 0.235 (0.130 - 0.423; P < 0.001); TP53wt: HR for DFS Gem vs. Obs, 0.794 (0.417 - 1.513; P = 0.483)] with a significant test for interaction (P = 0.003). In the TCGA dataset, TP53 mutations were associated with shortened DFS. CONCLUSIONS In CONKO-001, the benefit from adjuvant gemcitabine was confined to the TP53mut patient group. This potentially clinical relevant observation needs to be confirmed in independent prospective studies. The sensitivity of TP53mut PDAC to gemcitabine in CONKO-001 provides a lead for further mechanistic investigations.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/therapy
- Chemotherapy, Adjuvant/methods
- Clinical Trials, Phase III as Topic
- DNA Mutational Analysis
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Deoxycytidine/therapeutic use
- Disease-Free Survival
- Drug Resistance, Neoplasm/genetics
- Female
- High-Throughput Nucleotide Sequencing
- Humans
- Male
- Middle Aged
- Mutation
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/genetics
- Pancreatectomy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Prospective Studies
- Randomized Controlled Trials as Topic
- Tumor Suppressor Protein p53/genetics
- Gemcitabine
Collapse
Affiliation(s)
- Marianne Sinn
- Department of Medical Oncology and Haematology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- University Medical Center Hamburg-Eppendorf, Department of Oncology, Hematology and BMT, Hamburg, Germany
| | - Bruno V Sinn
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Denise Treue
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Central Biomaterial Bank Charité, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Frederik Damm
- Department of Medical Oncology and Haematology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Rosa Schmuck
- Department of General, Visceral and Transplantation Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, Germany
| | - Philipp Lohneis
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Frederick Klauschen
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, Germany
| | - Jana K Striefler
- Department of Medical Oncology and Haematology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Marcus Bahra
- Department of General, Visceral and Transplantation Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Hendrik Bläker
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sven Bischoff
- Department of Medical Oncology and Haematology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Uwe Pelzer
- Department of Medical Oncology and Haematology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Helmut Oettle
- Outpatient Department of Hematology/Oncology, Friedrichshafen, Germany
| | - Hanno Riess
- Department of Medical Oncology and Haematology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jan Budczies
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, Germany
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Carsten Denkert
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Institute of Pathology, Philipps-University Marburg and University Hospital Marburg (UKGM), Marburg, Germany
| |
Collapse
|
25
|
Duan H, Baratto L, Iagaru A. The Role of PET/CT in the Imaging of Pancreatic Neoplasms. Semin Ultrasound CT MR 2019; 40:500-508. [PMID: 31806148 DOI: 10.1053/j.sult.2019.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pancreas cancer is a complex disease and its prognosis is related to the origin of the tumor cell as well as the stage of disease at the time of diagnosis. Pancreatic adenocarcinomas derive from the exocrine pancreas and are the fourth leading cause of cancer-related deaths in the United States, while well-differentiated pancreatic neuroendocrine tumors (pNETs) derived from the endocrine part of the pancreas are rare and characterized by a slow growth and good life expectancy. Surgery is the only curative treatment approach, and an accurate assessment of resectability is of paramount importance in order to avoid futile procedures. The role of molecular imaging with positron emission tomography and computed tomography ranges from indispensable for pNETs to controversial for certain scenarios in pancreatic adenocarcinomas. This review article aims to overview molecular pancreatic imaging.
Collapse
Affiliation(s)
- Heying Duan
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA
| | - Lucia Baratto
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA.
| |
Collapse
|
26
|
Wang Z, Li Y, Zhan S, Zhang L, Zhang S, Tang Q, Li M, Tan Z, Liu S, Xing X. SMAD4 Y353C promotes the progression of PDAC. BMC Cancer 2019; 19:1037. [PMID: 31684910 PMCID: PMC6829834 DOI: 10.1186/s12885-019-6251-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 10/10/2019] [Indexed: 02/07/2023] Open
Abstract
Background SMAD4 is frequently inactivated and associated with a poor prognosis in pancreatic ductal adenocarcinoma (PDAC). Abnormal SMAD4 expression also plays an important role in the malignant progression of PDAC. Methods We investigated SMAD4 status in PDAC by immunohistochemical methods to explore the relationships between SMAD4 expression and clinicopathological features and then detected SMAD4 mutations by Sanger sequencing in 95 patients with PDAC to identify new mutation sites in PDAC. We further evaluated the effects of a missense mutation, Y353C, in the SMAD4 MH2 domain, on cell proliferation and migration in vitro. Results Immunohistochemistry showed that the expression of SMAD4 in PDAC carcinoma tissue was significantly lower than that in normal pancreatic tissue, and negative SMAD4 expression was closely related to tumour diameter, staging, lymph node metastasis and differentiation. Sanger sequencing analysis showed that the rate of SMAD4 mutation was 11.8% in 85 PDAC cases, and the novel SMAD4 Y353C missense mutation identified in this study promoted cell migration and invasion without affecting cell proliferation in vitro. Furthermore, SMAD4 Y353C resulted in reduced expression of E-cadherin and increased expression of Vimentin compared with wild-type SMAD4 overexpression. Conclusion This study supports the key role of SMAD4 as a tumour suppressor gene in PDAC and shows that SMAD4 Y353C is associated with poor progression of PDAC.
Collapse
Affiliation(s)
- Zusen Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yongxing Li
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shixiong Zhan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Zhang
- Prenatal Diagnosis Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Tang
- Prenatal Diagnosis Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Miaomiao Li
- Prenatal Diagnosis Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhen Tan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiguo Liu
- Prenatal Diagnosis Center, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Xiaoming Xing
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
27
|
Abstract
OBJECTIVES We evaluated how well phase II trials in locally advanced and metastatic pancreatic cancer (LAMPC) meet current recommendations for trial design. METHODS We conducted a systematic review of phase II first-line treatment trial for LAMPC. We assessed baseline characteristics, type of comparison, and primary end point to examine adherence to the National Cancer Institute recommendations for trial design. RESULTS We identified 148 studies (180 treatment arms, 7505 participants). Forty-seven (32%) studies adhered to none of the 5 evaluated National Cancer Institute recommendations, 62 (42%) followed 1, 31 (21%) followed 2, and 8 (5%) followed 3 recommendations. Studies varied with respect to the proportion of patients with good performance status (range, 0%-80%) and locally advanced disease (range, 14%-100%). Eighty-two (55%) studies concluded that investigational agents should progress to phase III testing; of these, 24 (16%) had documented phase III trials. Three (8%) phase III trials demonstrated clinically meaningful improvements for investigational agents. One of 38 phase II trials that investigated biological investigational agents was enriched for a biomarker. CONCLUSIONS Phase II trials do not conform well to current recommendations for trial design in LAMPC.
Collapse
|
28
|
Williet N, Saint A, Pointet AL, Tougeron D, Pernot S, Pozet A, Bechade D, Trouilloud I, Lourenco N, Hautefeuille V, Locher C, Desrame J, Artru P, Thirot Bidault A, Le Roy B, Pezet D, Phelip JM, Taieb J. Folfirinox versus gemcitabine/nab-paclitaxel as first-line therapy in patients with metastatic pancreatic cancer: a comparative propensity score study. Therap Adv Gastroenterol 2019; 12:1756284819878660. [PMID: 31598136 PMCID: PMC6764033 DOI: 10.1177/1756284819878660] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 08/16/2019] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Folfirinox (FFX) and gemcitabine/nab-paclitaxel (GN) are both standard first-line treatments in patients with metastatic pancreatic cancer (mPC). However, data comparing these two chemotherapeutic regimens and their sequential use remain scarce. METHODS Data from two independent cohorts enrolling patients treated with FFX (n = 107) or GN (n = 109) were retrospectively pooled. Primary endpoint was overall survival (OS). Progression-free survival (PFS) was the secondary endpoint. A propensity score based on age, gender, performance status (PS), and presence of liver metastases was used to make groups comparable. RESULTS In the whole study population, OS was significantly higher in FFX (14 months; 95% CI: 10-21) than in GN groups (9 months; 95% CI: 8-12) before (p = 0.008) and after (p = 0.021) adjusting for age, number of metastatic sites, liver metastases, peritoneal carcinomatosis and CA19.9 level at baseline. PFS tends to be higher in FFX (6 months) than GN groups (5 months; p = 0.053). After matching (n = 49/group), patients were comparable for all baseline characteristics including PS. In the matched population, there was a trend toward greater OS in patients treated with FFX (HR = 0.67; p = 0.097). However, survival in each group was not solely a result of the first-line regimen. The proportion of patients who were fit for GN after FFX failure (FFX-GN sequence) was higher (46.9%) than the reverse sequence (20.4%; p = 0.01), which suggests a higher feasibility for the FFX-GN sequence. Corresponding median OS were 19 months versus 9.5 months, respectively (p = 0.094). CONCLUSION This study shows greater OS with FFX than with GN in patients with mPC. GN after FFX failure appears more feasible than the reverse sequence.
Collapse
Affiliation(s)
| | - Angélique Saint
- Department of Medical Oncology, Antoine
Lacassagne Center, Nice, France
| | - Anne-Laure Pointet
- Department of Gastroenterology and
Gastro-intestinal Oncology, Hôpital Européen Georges-Pompidou, APHP, Paris
Descartes University, Sorbonne Paris Cité, Paris, France
| | - David Tougeron
- Department of Gastroenterology, Poitiers
University Hospital, Poitiers, France
| | - Simon Pernot
- Department of GI Oncology, Hôpital Européen
Georges-Pompidou, APHP, Paris Descartes University, Sorbonne Paris Cité,
Paris, France
| | - Astrid Pozet
- Methodology and Quality of Life in Oncology Unit
(INSERM UMR 1098), University Hospital of Besançon, Besançon, France
| | | | | | - Nelson Lourenco
- Gastroenterology Unit, Saint-Louis Teaching
Hospital, Paris, France
| | - Vincent Hautefeuille
- Department of Gastroenterology, Amiens-Picardie
University Hospital, Amiens, France
| | | | - Jérome Desrame
- Department of Oncology, Hôpital Privé Jean
Mermoz, Lyon, France
| | - Pascal Artru
- Department of Oncology, Hôpital Privé Jean
Mermoz, Lyon, France
| | - Anne Thirot Bidault
- Department of Gastroenterology, Hôpital Kremlin
Bicêtre, Le Kremlin-Bicêtre, Paris, France
| | - Bertrand Le Roy
- Department of Digestive and Hepatobiliary
Surgery, University Hospital of Saint-Etienne, Saint-Priest-en-Jarez,
France
| | - Denis Pezet
- Department of Digestive and Hepatobiliary
Surgery, University Hospital of Clermont-Ferrand, Clermont-Ferrand,
France
| | - Jean-Marc Phelip
- Department of Hepatogastroenterology,
University Hospital of Saint-Etienne, Saint-Etienne, France,EA 7425 HESPER, Health Services and Performance
Research, Claude Bernard Lyon 1 University, Lyon, France
| | - Julien Taieb
- Department of Gastroenterology and
Gastro-intestinal Oncology, Hôpital Européen Georges-Pompidou, APHP, Paris
Descartes University, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
29
|
Petrioli R, Torre P, Pesola G, Paganini G, Paolelli L, Miano ST, Martellucci I, Francini G, Francini E. Gemcitabine plus nab-paclitaxel followed by maintenance treatment with gemcitabine alone as first-line treatment for older adults with locally advanced or metastatic pancreatic cancer. J Geriatr Oncol 2019; 11:647-651. [PMID: 31471168 DOI: 10.1016/j.jgo.2019.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/20/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate the efficacy and safety of the combination Gemcitabine (Gem) plus nab-Paclitaxel (NabP) (Gem/NabP), followed by maintenance Gem in older adults with locally advanced or metastatic pancreatic cancer (PC). MATERIALS AND METHODS In this prospective observational study, the induction chemotherapy consisted of NabP 125 mg/m2 followed by Gem 1000 mg/m2 on days 1, 8, and 15 of a 4-week cycle. After a maximum of 3 cycles, patients without evidence of progressive disease (PD) were administered Gem 1000 mg/m2 weekly for 3 of 4 weeks as maintenance therapy until documentation of PD or unacceptable toxicity. The primary endpoint was six-month disease-control rate (DCR). RESULTS Overall, 36 patients >70 years with metastatic or locally advanced PC were enrolled at participating Institutions. After completion of Gem/NabP, 18 (50%) patients achieved partial response, 13 (36%) had stable disease, and 5 (14%) had PD. Thirty-one patients (86%) received Gem monotherapy as maintenance treatment for a median of 3 cycles (range, 2-9 cycles). Six-month DCR was 61% (95% CI, 45-77), median PFS was 6.4 months (95% CI, 5.4-8.3), and median OS was 13.4 months (95% CI, 11.1-16.7). During Gem/NabP regimen, the most common grade 3 toxicity included neutropenia (22%), anemia (19%) and thrombocytopenia (8%). Grade 3 neuropathy was not observed. During Gem maintenance therapy, grade 3 hematological toxicity was described in 6 patients (19%). CONCLUSION Gem/NabP followed by maintenance Gem appears to be safe and effective for older patients with locally advanced or metastatic PC.
Collapse
Affiliation(s)
- Roberto Petrioli
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy.
| | - Pamela Torre
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Guido Pesola
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Giovanni Paganini
- General Medicine& Oncology, Pieve di Coriano Hospital, ASST Mantova, Italy
| | | | - Salvatora Tindara Miano
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Ignazio Martellucci
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Guido Francini
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Edoardo Francini
- La Sapienza University, Rome, Italy; Medical Oncology Unit, Misericordia Hospital, Grosseto, Italy
| |
Collapse
|
30
|
|
31
|
Tahkola K, Leppänen J, Ahtiainen M, Väyrynen J, Haapasaari KM, Karttunen T, Kellokumpu I, Helminen O, Böhm J. Immune cell score in pancreatic cancer-comparison of hotspot and whole-section techniques. Virchows Arch 2019; 474:691-699. [PMID: 30843106 PMCID: PMC6581934 DOI: 10.1007/s00428-019-02549-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/28/2019] [Accepted: 02/21/2019] [Indexed: 01/04/2023]
Abstract
An immune cell score (ICS) was introduced for predicting survival in pancreatic ductal adenocarcinoma (PDAC). Few studies have compared different methods of evaluating immune infiltrate. This study compared ICSs determined in whole sections or tissue microarray-like hotspots for predicting survival after PDAC surgery. We included in 79 consecutive patients from a single geographical area that underwent surgery for PDAC (R0/R1, stages I-III). We performed digital image analyses to evaluate CD3 and CD8 staining. ICSs were classified as low, moderate, or high, based on the numbers of immune cells in the tumour core and invasive margin. We compared ICS groups determined with the hotspot and whole-section techniques. Associations between ICS and survival were analysed with Cox regression models, adjusted for sex, age, tumour stage, differentiation grade, perineural invasion, and resection radicality. In hotspot ICS analysis, 5-year overall survival rates for low, moderate, and high groups were 12.1%, 26.3%, and 26.8%, respectively (p = 0.193). In whole-section analyses, overall survival rates were 5.3%, 26.4%, and 43.8%, respectively (p = 0.030). In the adjusted Cox model, whole-section ICS groups were inversely associated with the overall mortality hazard ratio (HR): low, moderate, and high ICS groups had HRs of 1.00, 0.42 (95% CI 0.20-0.88), and 0.27 (95% CI 0.11-0.67), respectively. The number of immune cells per square millimetre in the tumour core and the invasive margin were significantly higher and had a wider range in hotspots than in whole-tissue sections. Accordingly, ICS could predict survival in patients with PDAC after surgery. Whole tissue section ICSs exhibited better prognostic value than hotspot ICSs.
Collapse
Affiliation(s)
- Kyösti Tahkola
- Department of Surgery, Central Finland Central Hospital, Jyvaskyla, Finland.
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland.
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland.
| | - Joni Leppänen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Maarit Ahtiainen
- Department of Education and Research, Central Finland Central Hospital and University of Eastern Finland, Jyvaskyla, Finland
| | - Juha Väyrynen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Kirsi-Maria Haapasaari
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Tuomo Karttunen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Ilmo Kellokumpu
- Department of Surgery, Central Finland Central Hospital, Jyvaskyla, Finland
| | - Olli Helminen
- Department of Surgery, Central Finland Central Hospital, Jyvaskyla, Finland
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Jan Böhm
- Department of Pathology, Central Finland Central Hospital, Jyvaskyla, Finland
| |
Collapse
|
32
|
Li Y, Feng Q, Jin J, Shi S, Zhang Z, Che X, Zhang J, Chen Y, Wu X, Chen R, Li S, Wang J, Li G, Li F, Dai M, Zheng L, Wang C. Experts’ consensus on intraoperative radiotherapy for pancreatic cancer. Cancer Lett 2019; 449:1-7. [DOI: 10.1016/j.canlet.2019.01.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/08/2019] [Accepted: 01/29/2019] [Indexed: 01/11/2023]
|
33
|
Consensus statement on mandatory measurements in pancreatic cancer trials (COMM-PACT) for systemic treatment of unresectable disease. Lancet Oncol 2019; 19:e151-e160. [PMID: 29508762 DOI: 10.1016/s1470-2045(18)30098-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/19/2017] [Accepted: 10/04/2017] [Indexed: 12/17/2022]
Abstract
Variations in the reporting of potentially confounding variables in studies investigating systemic treatments for unresectable pancreatic cancer pose challenges in drawing accurate comparisons between findings. In this Review, we establish the first international consensus on mandatory baseline and prognostic characteristics in future trials for the treatment of unresectable pancreatic cancer. We did a systematic literature search to find phase 3 trials investigating first-line systemic treatment for locally advanced or metastatic pancreatic cancer to identify baseline characteristics and prognostic variables. We created a structured overview showing the reporting frequencies of baseline characteristics and the prognostic relevance of identified variables. We used a modified Delphi panel of two rounds involving an international panel of 23 leading medical oncologists in the field of pancreatic cancer to develop a consensus on the various variables identified. In total, 39 randomised controlled trials that had data on 15 863 patients were included, of which 32 baseline characteristics and 26 prognostic characteristics were identified. After two consensus rounds, 23 baseline characteristics and 12 prognostic characteristics were designated as mandatory for future pancreatic cancer trials. The COnsensus statement on Mandatory Measurements in unresectable PAncreatic Cancer Trials (COMM-PACT) identifies a mandatory set of baseline and prognostic characteristics to allow adequate comparison of outcomes between pancreatic cancer studies.
Collapse
|
34
|
Glycosylation of ascites-derived exosomal CD133: a potential prognostic biomarker in patients with advanced pancreatic cancer. Med Mol Morphol 2019; 52:198-208. [DOI: 10.1007/s00795-019-00218-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/19/2019] [Indexed: 12/11/2022]
|
35
|
Nicoletti R, Chun R, Curran KM, Lorimier LPD, Morges MA, Rau S, Zwehlen CH, Thamm DH. Postsurgical Outcome in Cats with Exocrine Pancreatic Carcinoma: Nine Cases (2007–2016). J Am Anim Hosp Assoc 2018; 54:291-295. [DOI: 10.5326/jaaha-ms-6780] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
ABSTRACT
Feline exocrine pancreatic carcinoma has been reported to be an aggressive tumor with a high metastatic rate and poor prognosis. Studies reporting long-term outcome of cats after surgical removal of solitary pancreatic carcinomas are rare, due to the uncommon diagnosis and paucity of cats who undergo treatment. In this study, nine cases of feline exocrine pancreatic carcinoma from seven academic and private practice veterinary hospitals were reviewed to examine the outcome in cats undergoing surgical removal of the mass. The median postsurgical survival time for the nine cats was 316.5 days (range, 25–964 days), with three cats alive at a median follow-up time of 309 days. This study demonstrates that surgical removal of pancreatic exocrine tumors in cats with localized disease can result in survival times of over 300 days.
Collapse
Affiliation(s)
- Ryan Nicoletti
- From The Regional Veterinary Referral Center, Springfield, Virginia (R.N.); University of Wisconsin-Madison Veterinary Medical Teaching Hospital, Madison, Wisconsin (R.C.); Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon (K.M.C.); Centre Veterinaire Rive-Sud, Brossard, Quebec, Canada (L-P.d.L.); Red Bank Veterinary Hospital, Tinton Falls, New Jersey (M.A.M.); Me
| | - Ruthanne Chun
- From The Regional Veterinary Referral Center, Springfield, Virginia (R.N.); University of Wisconsin-Madison Veterinary Medical Teaching Hospital, Madison, Wisconsin (R.C.); Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon (K.M.C.); Centre Veterinaire Rive-Sud, Brossard, Quebec, Canada (L-P.d.L.); Red Bank Veterinary Hospital, Tinton Falls, New Jersey (M.A.M.); Me
| | - Kaitlin M. Curran
- From The Regional Veterinary Referral Center, Springfield, Virginia (R.N.); University of Wisconsin-Madison Veterinary Medical Teaching Hospital, Madison, Wisconsin (R.C.); Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon (K.M.C.); Centre Veterinaire Rive-Sud, Brossard, Quebec, Canada (L-P.d.L.); Red Bank Veterinary Hospital, Tinton Falls, New Jersey (M.A.M.); Me
| | - Louis-Philippe de Lorimier
- From The Regional Veterinary Referral Center, Springfield, Virginia (R.N.); University of Wisconsin-Madison Veterinary Medical Teaching Hospital, Madison, Wisconsin (R.C.); Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon (K.M.C.); Centre Veterinaire Rive-Sud, Brossard, Quebec, Canada (L-P.d.L.); Red Bank Veterinary Hospital, Tinton Falls, New Jersey (M.A.M.); Me
| | - Michelle A. Morges
- From The Regional Veterinary Referral Center, Springfield, Virginia (R.N.); University of Wisconsin-Madison Veterinary Medical Teaching Hospital, Madison, Wisconsin (R.C.); Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon (K.M.C.); Centre Veterinaire Rive-Sud, Brossard, Quebec, Canada (L-P.d.L.); Red Bank Veterinary Hospital, Tinton Falls, New Jersey (M.A.M.); Me
| | - Suzanne Rau
- From The Regional Veterinary Referral Center, Springfield, Virginia (R.N.); University of Wisconsin-Madison Veterinary Medical Teaching Hospital, Madison, Wisconsin (R.C.); Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon (K.M.C.); Centre Veterinaire Rive-Sud, Brossard, Quebec, Canada (L-P.d.L.); Red Bank Veterinary Hospital, Tinton Falls, New Jersey (M.A.M.); Me
| | - Courtney H. Zwehlen
- From The Regional Veterinary Referral Center, Springfield, Virginia (R.N.); University of Wisconsin-Madison Veterinary Medical Teaching Hospital, Madison, Wisconsin (R.C.); Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon (K.M.C.); Centre Veterinaire Rive-Sud, Brossard, Quebec, Canada (L-P.d.L.); Red Bank Veterinary Hospital, Tinton Falls, New Jersey (M.A.M.); Me
| | - Douglas H. Thamm
- From The Regional Veterinary Referral Center, Springfield, Virginia (R.N.); University of Wisconsin-Madison Veterinary Medical Teaching Hospital, Madison, Wisconsin (R.C.); Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon (K.M.C.); Centre Veterinaire Rive-Sud, Brossard, Quebec, Canada (L-P.d.L.); Red Bank Veterinary Hospital, Tinton Falls, New Jersey (M.A.M.); Me
| |
Collapse
|
36
|
Torres C, Grippo PJ. Pancreatic cancer subtypes: a roadmap for precision medicine. Ann Med 2018; 50:277-287. [PMID: 29537309 PMCID: PMC6151873 DOI: 10.1080/07853890.2018.1453168] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/25/2018] [Accepted: 03/09/2018] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is projected to become the second cause of cancer-related deaths by 2020. Although it has traditionally been approached as a disease, accumulated evidences point to the clinical heterogeneity of this disease, which translate into disparity in outcomes among the patients. Much emphasis has been put into patient classification introducing a platform for more tailored therapies. In the last 10 years, there have been important advances in the understanding of the molecular pathogenesis of PDAC, which has culminated with a comprehensive integrated genomic analysis from RNA expression profiles. Bailey et al. defined four subtypes and the different transcriptional networks underlying them. Firstly, we briefly describe and compare different subtyping approaches, which are mostly based on tissue mRNA expression analysis. We propose that these latest approaches to disease classification embrace not only those patients that are surgically resectable (20%), but even patients ineligible for surgery. Such considerations will include possible reclassification of these specific subtypes, enabling more personalized diagnosis and individualized treatment. The ultimate goal of this review is to identify current challenges in this area and summarize current efforts in developing clinical modalities that can effectively identify these subtypes in order to advance Precision Medicine. KEY MESSAGES • Pancreatic cancer can no longer be considered as one disease. • The heterogeneity underlying pancreatic cancer patients makes therapeutic options based on one-size-fits-all approach ineffective. • Identifying patients that could benefit from a specific treatment would help to avoid futile therapy approaches and to improve outcomes and quality of life of those whose long-term survival is unpromising.
Collapse
Affiliation(s)
- Carolina Torres
- a Department of Medicine , University of Illinois at Chicago , Chicago , IL , USA
| | - Paul J Grippo
- a Department of Medicine , University of Illinois at Chicago , Chicago , IL , USA
| |
Collapse
|
37
|
Wang Y, Fang T, Huang L, Wang H, Zhang L, Wang Z, Cui Y. Neutrophils infiltrating pancreatic ductal adenocarcinoma indicate higher malignancy and worse prognosis. Biochem Biophys Res Commun 2018; 501:313-319. [PMID: 29738769 DOI: 10.1016/j.bbrc.2018.05.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/04/2018] [Indexed: 02/07/2023]
Abstract
CD177 is considered to represent neutrophils. We analyzed mRNA expression level of CD177 and clinical follow-up survey of PDAC to estimate overall survival (OS) from Gene Expression Omnibus (GEO) dataset (GSE21501, containing samples from 102 PDAC patients) by R2 platform (http://r2.amc.nl). We also analyzed correlated genes of CD177 by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis to predict the potential relationship between neutrophils and prognosis of PDAC. We then performed hematoxylin and eosin (H&E) staining and immunohistochemical staining of surgical specimens to verify infiltration of neutrophils in PDAC tissues. After analyzing mRNA expression data and clinical follow-up survey provided in the GEO dataset (GSE21501, containing samples from 102 PDAC patients) and clinicopathological data of 23 PDAC patients, we demonstrated that CD177 was correlated with poor prognosis. The univariate Kaplan-Meier survival analysis revealed that OS was inversely associated with increased expression of CD177 (P = 0.012). Expression of phosphodiesterase (PDE)4D was positively related to CD177 in gene correlation analysis (R = 0.413, P < 0.001) by R2 platform. H&E staining and immunohistochemistry of CD177 in 23 PDAC surgical samples showed accumulation of neutrophils in the stroma and blood vessels around the cancer cells. In addition, immunohistochemical staining showed that CD177 was highly expressed in the stroma and blood vessels around tumor tissues of PDAC, which was similar to H&E staining. Expression of CD177 can be used to represent infiltration of neutrophils, which may have potential prognostic value in PDAC.
Collapse
Affiliation(s)
- Yufu Wang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang Province, China
| | - Tianyi Fang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang Province, China
| | - Lining Huang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang Province, China
| | - Hao Wang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang Province, China
| | - Lei Zhang
- Department of Pathology, Harbin Medical University, Harbin, 150000, Heilongjiang Province, China
| | - Zhidong Wang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang Province, China.
| | - Yunfu Cui
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang Province, China.
| |
Collapse
|
38
|
The Dutch Pancreas Biobank Within the Parelsnoer Institute: A Nationwide Biobank of Pancreatic and Periampullary Diseases. Pancreas 2018. [PMID: 29521943 DOI: 10.1097/mpa.0000000000001018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Large biobanks with uniform collection of biomaterials and associated clinical data are essential for translational research. The Netherlands has traditionally been well organized in multicenter clinical research on pancreatic diseases, including the nationwide multidisciplinary Dutch Pancreatic Cancer Group and Dutch Pancreatitis Study Group. To enable high-quality translational research on pancreatic and periampullary diseases, these groups established the Dutch Pancreas Biobank. METHODS The Dutch Pancreas Biobank is part of the Parelsnoer Institute and involves all 8 Dutch university medical centers and 5 nonacademic hospitals. Adult patients undergoing pancreatic surgery (all indications) are eligible for inclusion. Preoperative blood samples, tumor tissue from resected specimens, pancreatic cyst fluid, and follow-up blood samples are collected. Clinical parameters are collected in conjunction with the mandatory Dutch Pancreatic Cancer Audit. RESULTS Between January 2015 and May 2017, 488 patients were included in the first 5 participating centers: 4 university medical centers and 1 nonacademic hospital. Over 2500 samples were collected: 1308 preoperative blood samples, 864 tissue samples, and 366 follow-up blood samples. CONCLUSIONS Prospective collection of biomaterials and associated clinical data has started in the Dutch Pancreas Biobank. Subsequent translational research will aim to improve treatment decisions based on disease characteristics.
Collapse
|
39
|
Knudsen ES, Balaji U, Mannakee B, Vail P, Eslinger C, Moxom C, Mansour J, Witkiewicz AK. Pancreatic cancer cell lines as patient-derived avatars: genetic characterisation and functional utility. Gut 2018; 67:508-520. [PMID: 28073890 PMCID: PMC5868284 DOI: 10.1136/gutjnl-2016-313133] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) is a therapy recalcitrant disease with the worst survival rate of common solid tumours. Preclinical models that accurately reflect the genetic and biological diversity of PDAC will be important for delineating features of tumour biology and therapeutic vulnerabilities. DESIGN 27 primary PDAC tumours were employed for genetic analysis and development of tumour models. Tumour tissue was used for derivation of xenografts and cell lines. Exome sequencing was performed on the originating tumour and developed models. RNA sequencing, histological and functional analyses were employed to determine the relationship of the patient-derived models to clinical presentation of PDAC. RESULTS The cohort employed captured the genetic diversity of PDAC. From most cases, both cell lines and xenograft models were developed. Exome sequencing confirmed preservation of the primary tumour mutations in developed cell lines, which remained stable with extended passaging. The level of genetic conservation in the cell lines was comparable to that observed with patient-derived xenograft (PDX) models. Unlike historically established PDAC cancer cell lines, patient-derived models recapitulated the histological architecture of the primary tumour and exhibited metastatic spread similar to that observed clinically. Detailed genetic analyses of tumours and derived models revealed features of ex vivo evolution and the clonal architecture of PDAC. Functional analysis was used to elucidate therapeutic vulnerabilities of relevance to treatment of PDAC. CONCLUSIONS These data illustrate that with the appropriate methods it is possible to develop cell lines that maintain genetic features of PDAC. Such models serve as important substrates for analysing the significance of genetic variants and create a unique biorepository of annotated cell lines and xenografts that were established simultaneously from same primary tumour. These models can be used to infer genetic and empirically determined therapeutic sensitivities that would be germane to the patient.
Collapse
Affiliation(s)
- Erik S Knudsen
- University of Arizona Department of Medicine, University of Arizona, Tucson, Arizona, USA,University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA,McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Uthra Balaji
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Brian Mannakee
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Paris Vail
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Cody Eslinger
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher Moxom
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John Mansour
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Agnieszka K Witkiewicz
- University of Arizona Department of Medicine, University of Arizona, Tucson, Arizona, USA,University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA,McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA,University of Arizona Department of Pathology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
40
|
Awasthi N, Monahan S, Stefaniak A, Schwarz MA, Schwarz RE. Inhibition of the MEK/ERK pathway augments nab-paclitaxel-based chemotherapy effects in preclinical models of pancreatic cancer. Oncotarget 2017; 9:5274-5286. [PMID: 29435178 PMCID: PMC5797049 DOI: 10.18632/oncotarget.23684] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 12/19/2017] [Indexed: 01/05/2023] Open
Abstract
Nab-paclitaxel (NPT) combination with gemcitabine (Gem) represents the standard chemotherapy for pancreatic ductal adenocarcinoma (PDAC). Genetic alterations of the RAS/RAF/MEK/ERK (MAPK) signaling pathway yielding constitutive activation of the ERK cascade have been implicated as drivers of PDAC. Inhibition of downstream targets in the RAS-MAPK cascade such as MEK remains a promising therapeutic strategy. The efficacy of trametinib (Tra), a small molecule inhibitor of MEK1/2 kinase activity, in combination with nab-paclitaxel-based chemotherapy was evaluated in preclinical models of PDAC. The addition of trametinib to chemotherapy regimens showed a trend for an additive effect on tumor growth inhibition in subcutaneous AsPC-1 and Panc-1 PDAC xenografts. In a peritoneal dissemination model, median animal survival compared to controls (20 days) was increased after therapy with NPT (33 days, a 65% increase), Tra (31 days, a 55% increase), NPT+Tra (37 days, a 85% increase), NPT+Gem (39 days, a 95% increase) and NPT+Gem+Tra (49 days, a 145% increase). Effects of therapy on intratumoral proliferation and apoptosis corresponded with tumor growth inhibition. Trametinib effects were specifically accompanied by a decrease in phospho-ERK and an increase in cleaved caspase-3 and cleaved PARP-1 proteins. These findings suggest that the effects of nab-paclitaxel-based chemotherapy can be enhanced through specific inhibition of MEK1/2 kinase activity, and supports the clinical application of trametinib in combination with standard nab-paclitaxel-based chemotherapy in PDAC patients.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA.,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Sheena Monahan
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA
| | - Alexis Stefaniak
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Margaret A Schwarz
- Department of Pediatrics, Indiana University School of Medicine, South Bend, IN 46617, USA.,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Roderich E Schwarz
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA.,Goshen Center for Cancer Care, Goshen, IN 46526, USA
| |
Collapse
|
41
|
Lee B, Hutchinson R, Wong HL, Tie J, Putoczki T, Tran B, Gibbs P, Christie M. Emerging biomarkers for immunomodulatory cancer treatment of upper gastrointestinal, pancreatic and hepatic cancers. Semin Cancer Biol 2017; 52:241-252. [PMID: 29258858 DOI: 10.1016/j.semcancer.2017.12.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 12/14/2022]
Abstract
Carcinomas of the oesophagus, stomach, pancreas and liver are common and account for a disproportionately high number of cancer deaths. There is a need for new treatment options for patients with advanced disease. Immunomodulatory treatments including immune checkpoint blockade offer a promising new approach, with efficacy shown in other solid tumour types. However, only a small proportion of patients with carcinomas of the oesophagus, stomach, pancreas and liver have responded to single agent checkpoint inhibitors, and there is a need for markers that are predictive of response to guide treatment of individual patients. Predictive markers may include epidemiological factors such as ethnicity, the genomic status of the tumour, circulating markers, expression of immune checkpoint molecules, and other features of the stromal/immune response at the site of the tumour. This review will focus on predictive biomarkers for immune checkpoint blockade in oesophageal, gastric, pancreatic and hepatocellular carcinomas, including the genomic context and immune landscape in which they occur. Pancreatic carcinomas are largely resistant to immune checkpoint inhibition in trials to date, therefore emerging immunomodulatory treatments in this tumour type are also reviewed.
Collapse
Affiliation(s)
- Belinda Lee
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ryan Hutchinson
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Hui-Li Wong
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Jeanne Tie
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Tracy Putoczki
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ben Tran
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Peter Gibbs
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Michael Christie
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Pathology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| |
Collapse
|
42
|
Qiu L, Li H, Fu S, Chen X, Lu L. Surface markers of liver cancer stem cells and innovative targeted-therapy strategies for HCC. Oncol Lett 2017; 15:2039-2048. [PMID: 29434903 DOI: 10.3892/ol.2017.7568] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 11/02/2017] [Indexed: 12/20/2022] Open
Abstract
Liver cancer stem cells (LCSCs) have important roles in the occurrence, development, recurrence, therapy resistance and metastasis of hepatocellular carcinoma (HCC). Therefore, intensive studies are undergoing to identify the mechanisms by which LCSCs contribute to HCC invasion and metastasis, and to design more efficient treatments for this disease. With continuous efforts in LCSC research over the years, therapies targeting LCSCs are thought to have great potential for the clinical treatment and prognosis of liver cancer. Novel LCSC surface markers are continuously discovered and several have been used in targeted therapies to reduce HCC recurrence, metastasis, and drug resistance following tumor resection. The present review describes the surface markers characterizing LCSCs and the recent progress in therapies targeting these markers, including antibodies and polypeptides.
Collapse
Affiliation(s)
- Lige Qiu
- Department of Intervention, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Hailiang Li
- Department of Intervention, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China.,Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Sirui Fu
- Department of Intervention, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Xiaofang Chen
- Department of Intervention, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China.,Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China.,Stem Cell and Regenerative Medicine Laboratory, Beijing Institute of Transfusion Medicine, Beijing 100850, P.R. China
| | - Ligong Lu
- Department of Intervention, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, P.R. China
| |
Collapse
|
43
|
Varadhachary GR, Wolff RA. Current and Evolving Therapies for Metastatic Pancreatic Cancer: Are We Stuck With Cytotoxic Chemotherapy? J Oncol Pract 2017; 12:797-805. [PMID: 27621332 DOI: 10.1200/jop.2016.015586] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
At present, front-line therapy for metastatic pancreatic ductal adenocarcinoma is combination chemotherapy, most commonly FOLFIRINOX (fluorouracil, irinotecan, and oxaliplatin) or gemcitabine and nanoparticle albumin-bound paclitaxel. Despite a better understanding of the genomic landscape and the importance of the tumor microenvironment, we have not made a seismic shift in the overall survival for this disease. Given our growing understanding of the biology of pancreatic ductal adenocarcinoma, the question remains whether novel, noncytotoxic agents will augment or even replace conventional chemotherapy. The thrust of ongoing efforts can be divided into broad categories, including exploiting the DNA damage repair phenotype, stroma and specific pathway-targeting agents, and enhancing immune destruction of pancreatic ductal adenocarcinoma. In this article, we review the current and evolving therapeutic landscape of metastatic pancreatic cancer.
Collapse
Affiliation(s)
| | - Robert A Wolff
- The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
44
|
Moravec R, Divi R, Verma M. Detecting circulating tumor material and digital pathology imaging during pancreatic cancer progression. World J Gastrointest Oncol 2017; 9:235-250. [PMID: 28656074 PMCID: PMC5472554 DOI: 10.4251/wjgo.v9.i6.235] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/04/2017] [Accepted: 03/24/2017] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer (PC) is a leading cause of cancer-related death worldwide. Clinical symptoms typically present late when treatment options are limited and survival expectancy is very short. Metastatic mutations are heterogeneous and can accumulate up to twenty years before PC diagnosis. Given such genetic diversity, detecting and managing the complex states of disease progression may be limited to imaging modalities and markers present in circulation. Recent developments in digital pathology imaging show potential for early PC detection, making a differential diagnosis, and predicting treatment sensitivity leading to long-term survival in advanced stage patients. Despite large research efforts, the only serum marker currently approved for clinical use is CA 19-9. Utility of CA 19-9 has been shown to improve when it is used in combination with PC-specific markers. Efforts are being made to develop early-screening assays that can detect tumor-derived material, present in circulation, before metastasis takes a significant course. Detection of markers that identify circulating tumor cells and tumor-derived extracellular vesicles (EVs) in biofluid samples offers a promising non-invasive method for this purpose. Circulating tumor cells exhibit varying expression of epithelial and mesenchymal markers depending on the state of tumor differentiation. This offers a possibility for monitoring disease progression using minimally invasive procedures. EVs also offer the benefit of detecting molecular cargo of tumor origin and add the potential to detect circulating vesicle markers from tumors that lack invasive properties. This review integrates recent genetic insights of PC progression with developments in digital pathology and early detection of tumor-derived circulating material.
Collapse
|
45
|
Wang L, Li P, Hu W, Xia Y, Hu C, Liu L, Jiang X. CD44 +CD24 + subset of PANC-1 cells exhibits radiation resistance via decreased levels of reactive oxygen species. Oncol Lett 2017; 14:1341-1346. [PMID: 28789349 PMCID: PMC5529798 DOI: 10.3892/ol.2017.6301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 02/28/2017] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence has suggested that pancreatic adenocarcinoma is sustained by pancreatic cancer stem cells. The present study aimed to investigate the expression patterns of the pancreatic cancer stem cell surface markers cluster of differentiation CD44 and CD24 in a pancreatic adenocarcinoma cell line, and to investigate the possible mechanisms for their radiation resistance. Flow cytometry was used to analyze the expression patterns of CD44 and CD24 in the pancreatic adenocarcinoma PANC-1 cell line. In addition, a multi-target click model was used to fit cell survival curves and determine the sensitizer enhancement ratio. The apoptosis and cycle distribution of the four cell subsets was determined using flow cytometry, and the level of reactive oxygen species (ROS) was determined using the 2',7'-dichlorofluorescin diacetate probe. The present results identified that the ratios of CD44+ and CD24+ in the sorted PANC-1 cell line were 92.0 and 4.7%, respectively. Prior to radiation, no statistically significant differences were observed among the four groups. Following treatment with 6 MV of X-rays, the rate of apoptosis was decreased in the CD44+CD24+ group compared with other subsets. The percentage of G0/G1 cells was highest in the CD44+CD24+ group compared with the three other groups, which exhibited increased radiosensitivity. In addition, the level of ROS in the CD44+CD24+ group was reduced compared with the other groups. In summary, the results of the present study indicated that CD44+CD24+ exhibited stem cell properties. The lower level of ROS and apoptosis in CD44+CD24+ cells may contribute to their resistance to radiation in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Lei Wang
- Department of Radiation Oncology, Lianyungang First People's Hospital, Lianyugang, Jiangsu 222002, P.R. China
| | - Pengping Li
- Department of Bioinformatics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Hu
- Department of Hepatobiliary Surgery, Lianyungang First People's Hospital, Lianyungang, Jiangsu 222002, P.R. China
| | - Youyou Xia
- Department of Radiation Oncology, Lianyungang First People's Hospital, Lianyugang, Jiangsu 222002, P.R. China
| | - Chenxi Hu
- Department of Radiation Oncology, Lianyungang First People's Hospital, Lianyugang, Jiangsu 222002, P.R. China
| | - Liang Liu
- Department of Radiation Oncology, Lianyungang First People's Hospital, Lianyugang, Jiangsu 222002, P.R. China
| | - Xiaodong Jiang
- Department of Radiation Oncology, Lianyungang First People's Hospital, Lianyugang, Jiangsu 222002, P.R. China
| |
Collapse
|
46
|
Hamada C, Okusaka T, Ikari T, Isayama H, Furuse J, Ishii H, Nakai Y, Imai S, Okamura S. Efficacy and safety of gemcitabine plus S-1 in pancreatic cancer: a pooled analysis of individual patient data. Br J Cancer 2017; 116:1544-1550. [PMID: 28472821 PMCID: PMC5518857 DOI: 10.1038/bjc.2017.128] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 03/29/2017] [Accepted: 04/12/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Three randomised trials (GEST, JACCRO PC-01, and GEMSAP) were conducted to evaluate the efficacy of gemcitabine plus S-1 (GS) vs gemcitabine alone in patients with advanced pancreatic cancer (PC). In this pooled analysis, the efficacy and safety of GS vs gemcitabine were evaluated. METHODS Additional follow-up was conducted and survival data were updated in each study. A total of 770 patients (gemcitabine 389; GS 381) were included in the pooled analysis. The efficacy and safety data were analysed according to disease extent: locally advanced PC (LAPC) or metastatic PC (MPC). RESULTS There were 738 (95.8%) overall survival events. In patients with LAPC (n=193), the median survival was 11.83 months for gemcitabine and 16.41 months for GS (hazard ratio (HR)=0.708; 95% confidence intervals (CI), 0.527-0.951; P=0.0220). In patients with MPC (n=577), the median survival was 8.02 months for gemcitabine and 9.43 months for GS (HR=0.872; 95% CI, 0.738-1.032; P=0.1102). The rate of grade 3/4 toxicity (rash and thrombocytopenia in LAPC; rash, diarrhoea, vomiting, and neutropaenia in MPC) was significantly higher for GS than for gemcitabine. CONCLUSIONS Gemcitabine plus S-1 is a viable treatment alternative to gemcitabine, which is one of the standard treatments in patients with LAPC.
Collapse
Affiliation(s)
- Chikuma Hamada
- Department of Management Science, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Takaaki Ikari
- Department of Internal Medicine, Tobu Chiiki Hospital, 5-14-1 Kameari, Katsushika-ku, Tokyo 125-8512, Japan
| | - Hiroyuki Isayama
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo 181-8611, Japan
| | - Hiroshi Ishii
- Department of Medical Oncology, Shikoku Cancer Center, 160 Kou, Minami Umemoto, Matsuyama 791-0280, Japan
| | - Yousuke Nakai
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Shogo Imai
- Department of Management Science, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Shota Okamura
- Department of Management Science, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan
| |
Collapse
|
47
|
Lai X, Wang M, McElyea SD, Sherman S, House M, Korc M. A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer. Cancer Lett 2017; 393:86-93. [PMID: 28232049 PMCID: PMC5386003 DOI: 10.1016/j.canlet.2017.02.019] [Citation(s) in RCA: 242] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 02/13/2017] [Accepted: 02/13/2017] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly malignancy that often presents clinically at an advanced stage and that may be confused with chronic pancreatitis (CP). Conversely, CP may be misdiagnosed as PDAC leading to unwarranted pancreas resection. Therefore, early PDAC diagnosis and clear differentiation between PDAC and CP are crucial for improved care. Exosomes are circulating microvesicles whose components can serve as cancer biomarkers. We compared exosomal glypican-1 (GPC1) and microRNA levels in normal control subjects and in patients with PDAC and CP. We report that exosomal GPC1 is not diagnostic for PDAC, whereas high exosomal levels of microRNA-10b, (miR-10b), miR-21, miR-30c, and miR-181a and low miR-let7a readily differentiate PDAC from normal control and CP samples. By contrast with GPC1, elevated exosomal miR levels decreased to normal values within 24 h following PDAC resection. All 29 PDAC cases exhibited significantly elevated exosomal miR-10b and miR-30c levels, whereas 8 cases had normal or slightly increased CA 19-9 levels. Thus, our exosomal miR signature is superior to exosomal GPC1 or plasma CA 19-9 levels in establishing a diagnosis of PDAC and differentiating between PDAC and CP.
Collapse
MESH Headings
- Area Under Curve
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- CA-19-9 Antigen/blood
- Carcinoma, Pancreatic Ductal/blood
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/surgery
- Case-Control Studies
- Chromatography, Liquid
- Diagnosis, Differential
- Exosomes/metabolism
- Gene Expression Profiling/methods
- Glypicans/blood
- Humans
- MicroRNAs/blood
- MicroRNAs/genetics
- Oligonucleotide Array Sequence Analysis
- Pancreatic Neoplasms/blood
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/surgery
- Pancreatitis, Chronic/blood
- Pancreatitis, Chronic/diagnosis
- Pancreatitis, Chronic/genetics
- Predictive Value of Tests
- ROC Curve
- Reproducibility of Results
- Tandem Mass Spectrometry
- Transcriptome
Collapse
Affiliation(s)
- Xianyin Lai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Mu Wang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Samantha Deitz McElyea
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stuart Sherman
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Pancreatic Cancer Signature Center, Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Michael House
- Pancreatic Cancer Signature Center, Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA; Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Murray Korc
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Pancreatic Cancer Signature Center, Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA.
| |
Collapse
|
48
|
Hagiwara Y, Ohashi Y, Okusaka T, Ueno H, Ioka T, Boku N, Egawa S, Hatori T, Furuse J, Mizumoto K, Ohkawa S, Yamaguchi T, Yamao K, Funakoshi A, Cheng AL, Kihara K, Sato A, Tanaka M. Health-related quality of life in a randomised phase III study of gemcitabine plus S-1, S-1 alone and gemcitabine alone for locally advanced or metastatic pancreatic cancer: GEST study. ESMO Open 2017; 2:e000151. [PMID: 28761731 PMCID: PMC5519786 DOI: 10.1136/esmoopen-2016-000151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 12/14/2022] Open
Abstract
Objective: This study was performed to compare health-related quality of life (HRQOL) of gemcitabine plus S-1 (GS), S-1 alone and gemcitabine alone as first-line chemotherapy for locally advanced or metastatic pancreatic cancer in the GEST (Gemcitabine and TS-1 Trial) study and to assess the impacts of adverse events and tumour response on HRQOL. Methods: Patients were randomly assigned to receive gemcitabine alone (1000 mg/m2 weekly for 3 of 4 weeks), S-1 alone (80, 100 or 120 mg/day twice daily for 4 of 6 weeks) or GS (gemcitabine at 1000 mg/m2 weekly plus S-1 at 60, 80 or 100 mg/day twice daily for 2 of 3 weeks). HRQOL was assessed using the EuroQoL-5D (EQ-5D) questionnaire at baseline and weeks 6, 12, 24, 48 and 72. EQ-5D scores, quality-adjusted life months (QALMs), quality-adjusted progression-free months (QAPFMs) and time until definitive HRQOL deterioration (TUDD) were compared among the three groups. The impacts of adverse events and tumour response on EQ-5D scores were analysed. Results: Including EQ-5D scores after death as 0, the mean profile was significantly better in the GS than gemcitabine group (difference, 0.069; p=0.003), but not the S-1 group (difference, −0.011; p=0.613). The mean profiles until death were similar in the three groups. QALMs, QAPFMs and TUDD were significantly longer in the GS than gemcitabine group (p<0.001, p<0.001 and p=0.004, respectively), but not the S-1 group (p=0.563, p=0.741 and p=0.701, respectively). Fatigue, anorexia and tumour response were significantly associated with changes in EQ-5D scores. Conclusions: GS achieved better HRQOL than gemcitabine alone, resulting a good balance between overall survival and HRQOL benefits. S-1 alone provides HRQOL similar to that provided by gemcitabine alone. Preventing fatigue and anorexia and maintaining better response would improve HRQOL.
Collapse
Affiliation(s)
| | - Yasuo Ohashi
- Department of Integrated Science and Engineering for Sustainable Society, Chuo University, Tokyo, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hideki Ueno
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsuya Ioka
- Department of Hepatobiliary and Pancreatic Oncology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Narikazu Boku
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | | | - Takashi Hatori
- Digestive Diseases Center, International University of Health and Welfare Mita Hospital, Tokyo, Japan
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University School of Medicine, Tokyo, Japan
| | | | - Shinichi Ohkawa
- Division of Hepatobiliary and Pancreatic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Taketo Yamaguchi
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Kenji Yamao
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | | | - Ann-Lii Cheng
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Kiyohiro Kihara
- Department of Biostatistics, The University of Tokyo, Tokyo, Japan
| | - Atsushi Sato
- Department of Medical Oncology, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Masao Tanaka
- Department of Surgery, Shimonoseki City Hospital, Shimonoseki, Japan
| |
Collapse
|
49
|
Preclinical Rationale for the Phase III Trials in Metastatic Pancreatic Cancer: Is Wishful Thinking Clouding Successful Drug Development for Pancreatic Cancer? Pancreas 2017; 46:143-150. [PMID: 28085753 PMCID: PMC5242389 DOI: 10.1097/mpa.0000000000000753] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prior phase III trials in advanced pancreatic cancer have been predominantly unsuccessful. In this review, we attempt to understand how past preclinical data were translated into phase III clinical trials in metastatic pancreatic cancer as described in the article. A systematic literature review conducted through the MEDLINE, Embase, and Cochrane Central Register of Controlled Trials databases, from January 1997 to June 2015 using key words-phase III clinical trials, metastatic/advanced pancreatic adenocarcinoma or pancreatic cancer identified 30 randomized controlled trials (RCTs) that met criteria. The trials were limited to RCTs in the first-line treatment of patients with metastatic pancreatic cancer. The success rate of first-line phase III studies in advanced pancreatic cancer was only 13%. In 60% of the RCTs, no preclinical experiments were referenced in biologically cognate pancreatic models. Nine (30%) of the RCTs were designed based on preclinical evidence from in vitro cell lines alone without additional in vivo validation in xenograft models. It remains uncertain how strongly the preclinical data influence the development of clinical regimens but so far the studies developed based on more solid preclinical evidence have been successful.
Collapse
|
50
|
Matrisian LM, Berlin JD. The Past, Present, and Future of Pancreatic Cancer Clinical Trials. Am Soc Clin Oncol Educ Book 2017; 35:e205-15. [PMID: 27249725 DOI: 10.1200/edbk_159117] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Upper gastrointestinal malignancies comprise half of the deadliest cancers as defined by those with a 5-year survival rate less than 50%. Using pancreatic adenocarcinoma (PAC) as an example, we retrospectively evaluated the success of phase III clinical trials, examined the current landscape of clinical trials, and identified emerging areas that foretell the future for this disease. Pancreatic and liver cancers are on the rise and will be the second and third leading causes of cancer deaths in 2030. A total of 35 different agents or combinations have been tested in randomized phase III clinical trials for patients with advanced PAC over the past 25 years, but only 11% have been incorporated into clinical practice. There has been a 37% increase in the number of PAC trials open in the United States between 2011 to 2012 and 2014 to 2015. Enrollment has also increased slightly, from 3.85% of the newly diagnosed cases in 2011 to 4.15% in 2014. However, the demand for patients far exceeds the number of patients available for these trials. On the horizon is the realization that stratification of patients with PAC using biomarkers that predict a high probability of a response could reallocate patients to faster, smaller trials with a greater chance of a survival benefit. The current landscape of PAC clinical trials and the launch of the Pancreatic Cancer Action Network's Know Your Tumor initiative indicate this shift is starting to occur, with particular emphasis on targeted therapies, immunotherapies, and agents that disrupt the stroma.
Collapse
Affiliation(s)
- Lynn M Matrisian
- From the Pancreatic Cancer Action Network, Manhattan Beach, CA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN
| | - Jordan D Berlin
- From the Pancreatic Cancer Action Network, Manhattan Beach, CA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN
| |
Collapse
|