1
|
Choi D, Gonzalez‐Suarez AM, Dumbrava MG, Medlyn M, de Hoyos‐Vega JM, Cichocki F, Miller JS, Ding L, Zhu M, Stybayeva G, Gaspar‐Maia A, Billadeau DD, Ma WW, Revzin A. Microfluidic Organoid Cultures Derived from Pancreatic Cancer Biopsies for Personalized Testing of Chemotherapy and Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303088. [PMID: 38018486 PMCID: PMC10837378 DOI: 10.1002/advs.202303088] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/17/2023] [Indexed: 11/30/2023]
Abstract
Patient-derived cancer organoids (PDOs) hold considerable promise for personalizing therapy selection and improving patient outcomes. However, it is challenging to generate PDOs in sufficient numbers to test therapies in standard culture platforms. This challenge is particularly acute for pancreatic ductal adenocarcinoma (PDAC) where most patients are diagnosed at an advanced stage with non-resectable tumors and where patient tissue is in the form of needle biopsies. Here the development and characterization of microfluidic devices for testing therapies using a limited amount of tissue or PDOs available from PDAC biopsies is described. It is demonstrated that microfluidic PDOs are phenotypically and genotypically similar to the gold-standard Matrigel organoids with the advantages of 1) spheroid uniformity, 2) minimal cell number requirement, and 3) not relying on Matrigel. The utility of microfluidic PDOs is proven by testing PDO responses to several chemotherapies, including an inhibitor of glycogen synthase kinase (GSKI). In addition, microfluidic organoid cultures are used to test effectiveness of immunotherapy comprised of NK cells in combination with a novel biologic. In summary, our microfluidic device offers considerable benefits for personalizing oncology based on cancer biopsies and may, in the future, be developed into a companion diagnostic for chemotherapy or immunotherapy treatments.
Collapse
Affiliation(s)
- Daheui Choi
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMN55905USA
| | | | - Mihai G. Dumbrava
- Division of Experimental PathologyMayo ClinicRochesterMN55905USA
- Center for Individualized MedicineEpigenomics programMayo ClinicRochesterMN55905USA
| | - Michael Medlyn
- Division of Oncology ResearchCollege of MedicineMayo ClinicRochesterMN55905USA
| | | | - Frank Cichocki
- Department of MedicineUniversity of MinnesotaMinneapolisMN55455USA
| | | | - Li Ding
- Division of Oncology ResearchCollege of MedicineMayo ClinicRochesterMN55905USA
| | - Mojun Zhu
- Division of Medical OncologyMayo ClinicRochesterMN55905USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMN55905USA
| | - Alexandre Gaspar‐Maia
- Division of Experimental PathologyMayo ClinicRochesterMN55905USA
- Center for Individualized MedicineEpigenomics programMayo ClinicRochesterMN55905USA
| | - Daniel D. Billadeau
- Division of Oncology ResearchCollege of MedicineMayo ClinicRochesterMN55905USA
| | - Wen Wee Ma
- Division of Medical OncologyMayo ClinicRochesterMN55905USA
| | - Alexander Revzin
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMN55905USA
| |
Collapse
|
2
|
Ziogas DC, Papadopoulou E, Gogas H, Sakellariou S, Felekouras E, Theocharopoulos C, Stefanou DT, Theochari M, Boukovinas I, Matthaios D, Koumarianou A, Zairi E, Liontos M, Koutsoukos K, Metaxa-Mariatou V, Kapetsis G, Meintani A, Tsaousis GN, Nasioulas G. Digging into the NGS Information from a Large-Scale South European Population with Metastatic/Unresectable Pancreatic Ductal Adenocarcinoma: A Real-World Genomic Depiction. Cancers (Basel) 2023; 16:2. [PMID: 38201431 PMCID: PMC10778112 DOI: 10.3390/cancers16010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024] Open
Abstract
Despite ongoing oncological advances, pancreatic ductal adenocarcinoma (PDAC) continues to have an extremely poor prognosis with limited targeted and immunotherapeutic options. Its genomic background has not been fully characterized yet in large-scale populations all over the world. Methods: Replicating a recent study from China, we collected tissue samples from consecutive Greek patients with pathologically-confirmed metastatic/unresectable PDAC and retrospectively investigated their genomic landscape using next generation sequencing (NGS). Findings: From a cohort of 409 patients, NGS analysis was successfully achieved in 400 cases (56.50% males, median age: 61.8 years). Consistent with a previous study, KRAS was the most frequently mutated gene in 81.50% of tested samples, followed by TP53 (50.75%), CDKN2 (8%), and SMAD4 (7.50%). BRCA1/2 variants with on-label indications were detected in 2%, and 87.50% carried a variant associated with off-label treatment (KRAS, ERBB2, STK11, or HRR-genes), while 3.5% of the alterations had unknown/preliminary-studied actionability (TP53/CDKN2A). Most of HRR-alterations were in intermediate- and low-risk genes (CHEK2, RAD50, RAD51, ATM, FANCA, FANCL, FANCC, BAP1), with controversial actionability: 8% harbored a somatic non-BRCA1/2 alteration, 6 cases had a high-risk alteration (PALB2, RAD51C), and one co-presented a PALB2/BRCA2 alteration. Elevated LOH was associated with HRR-mutated status and TP53 mutations while lowered LOH was associated with KRAS alterations. Including TMB/MSI data, the potential benefit from an NGS-oriented treatment was increased from 1.91% to 13.74% (high-MSI: 0.3%, TMB > 10 muts/MB: 12.78%). TMB was slightly increased in females (4.75 vs. 4.46 muts/MB) and in individuals with age > 60 (4.77 vs. 4.40 muts/MB). About 28.41% showed PD-L1 > 1% either in tumor or immune cells, 15.75% expressed PD-L1 ≥ 10%, and only 1.18% had PD-L1 ≥ 50%. This is the largest depiction of real-world genomic characteristics of European patients with PDAC, which offers some useful clinical and research insights.
Collapse
Affiliation(s)
- Dimitrios C. Ziogas
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (H.G.); (C.T.); (D.T.S.); (M.T.)
| | - Eirini Papadopoulou
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| | - Helen Gogas
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (H.G.); (C.T.); (D.T.S.); (M.T.)
| | - Stratigoula Sakellariou
- First Department of Pathology, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Evangellos Felekouras
- First Department of Surgery, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Charalampos Theocharopoulos
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (H.G.); (C.T.); (D.T.S.); (M.T.)
| | - Dimitra T. Stefanou
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (H.G.); (C.T.); (D.T.S.); (M.T.)
| | - Maria Theochari
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (H.G.); (C.T.); (D.T.S.); (M.T.)
| | - Ioannis Boukovinas
- Department of Medical Oncology, Bioclinic Hospital, 54622 Thessaloniki, Greece;
| | | | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Eleni Zairi
- Oncology Department, St. Lukes Hospital, 55236 Thessaloniki, Greece;
| | - Michalis Liontos
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (M.L.); (K.K.)
| | - Konstantinos Koutsoukos
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; (M.L.); (K.K.)
| | - Vasiliki Metaxa-Mariatou
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| | - George Kapetsis
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| | - Angeliki Meintani
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| | - Georgios N. Tsaousis
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| | - George Nasioulas
- GeneKor Medical S.A., 15344 Gerakas, Greece; (E.P.); (V.M.-M.); (G.K.); (A.M.); (G.N.T.); (G.N.)
| |
Collapse
|
3
|
Ibrahim R, Khoury R, Ibrahim T, Assi T, Cesne AL. KRAS G12C mutation: from black sheep to key player in pancreatic cancer treatment. Future Oncol 2023; 19:485-488. [PMID: 36946253 DOI: 10.2217/fon-2023-0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Affiliation(s)
- Rebecca Ibrahim
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France
| | - Rita Khoury
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France
| | - Tony Ibrahim
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France
| | - Tarek Assi
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France
| | - Axel Le Cesne
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
4
|
Khoury R, Ibrahim R, Ibrahim T, Cesne AL, Assi T. NALIRIFOX in the upfront setting of metastatic pancreatic cancer: can FOLFIRINOX finally be dethroned? Future Oncol 2023; 19:617-620. [PMID: 37129008 DOI: 10.2217/fon-2023-0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Affiliation(s)
- Rita Khoury
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, 94800, France
| | - Rebecca Ibrahim
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, 94800, France
| | - Tony Ibrahim
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, 94800, France
| | - Axel Le Cesne
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, 94800, France
| | - Tarek Assi
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, 94800, France
| |
Collapse
|
5
|
de Almeida DRQ, Dos Santos AF, Wailemann RAM, Terra LF, Gomes VM, Arini GS, Bertoldi ERM, Reis EM, Baptista MS, Labriola L. Necroptosis activation is associated with greater methylene blue-photodynamic therapy-induced cytotoxicity in human pancreatic ductal adenocarcinoma cells. Photochem Photobiol Sci 2022; 22:729-744. [PMID: 36495407 DOI: 10.1007/s43630-022-00347-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinomas (PDAC) are the fourth leading cause of death due to neoplasms. In view of the urgent need of effective treatments for PDAC, photodynamic therapy (PDT) appears as a promising alternative. However, its efficacy against PDAC and the mechanisms involved in cell death induction remain unclear. In this study, we set out to evaluate PDT's cytotoxicity using methylene blue (MB) as a photosensitizer (PS) (MB-PDT) and to evaluate the contribution of necroptosis in its effect in human PDAC cells. Our results demonstrated that MB-PDT induced significant death of different human PDAC models presenting two different susceptibility profiles. This effect was independent of MB uptake or its subcellular localization. We found that the ability of triggering necroptosis was determinant to increase the treatment efficiency. Analysis of single cell RNA-seq data from normal and neoplastic human pancreatic tissues showed that specific necroptosis proteins RIPK1, RIPK3 and MLKL presented significant higher expression levels in cells displaying a transformed phenotype providing further support to the use of approaches that activate necroptosis, like MB-PDT, as useful adjunct to surgery of PDAC to tackle the problem of microscopic residual disease as well as to minimize the chance of local and metastatic recurrence.
Collapse
Affiliation(s)
- Daria R Q de Almeida
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Cidade Universitária, Block 09, Room 976, Av. Professor Lineu Prestes 748, São Paulo, 05508-000, Brazil
| | - Ancély F Dos Santos
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Cidade Universitária, Block 09, Room 976, Av. Professor Lineu Prestes 748, São Paulo, 05508-000, Brazil
| | - Rosangela A M Wailemann
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Cidade Universitária, Block 09, Room 976, Av. Professor Lineu Prestes 748, São Paulo, 05508-000, Brazil
| | - Letícia F Terra
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Cidade Universitária, Block 09, Room 976, Av. Professor Lineu Prestes 748, São Paulo, 05508-000, Brazil
| | - Vinícius M Gomes
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Cidade Universitária, Block 09, Room 976, Av. Professor Lineu Prestes 748, São Paulo, 05508-000, Brazil
| | - Gabriel S Arini
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Cidade Universitária, Block 09, Room 976, Av. Professor Lineu Prestes 748, São Paulo, 05508-000, Brazil
| | - Ester R M Bertoldi
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Cidade Universitária, Block 09, Room 976, Av. Professor Lineu Prestes 748, São Paulo, 05508-000, Brazil
| | - Eduardo M Reis
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Cidade Universitária, Block 09, Room 976, Av. Professor Lineu Prestes 748, São Paulo, 05508-000, Brazil
| | - Maurício S Baptista
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Cidade Universitária, Block 09, Room 976, Av. Professor Lineu Prestes 748, São Paulo, 05508-000, Brazil.
| | - Leticia Labriola
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Cidade Universitária, Block 09, Room 976, Av. Professor Lineu Prestes 748, São Paulo, 05508-000, Brazil.
| |
Collapse
|
6
|
Liew HS, Mai CW, Zulkefeli M, Madheswaran T, Kiew LV, Pua LJW, Hii LW, Lim WM, Low ML. Novel Gemcitabine-Re(I) Bisquinolinyl Complex Combinations and Formulations With Liquid Crystalline Nanoparticles for Pancreatic Cancer Photodynamic Therapy. Front Pharmacol 2022; 13:903210. [PMID: 35873548 PMCID: PMC9299370 DOI: 10.3389/fphar.2022.903210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/17/2022] [Indexed: 12/24/2022] Open
Abstract
With less than 10% of 5-year survival rate, pancreatic ductal adenocarcinoma (PDAC) is known to be one of the most lethal types of cancer. Current literature supports that gemcitabine is the first-line treatment of PDAC. However, poor cellular penetration of gemcitabine along with the acquired and intrinsic chemoresistance of tumor against it often reduced its efficacy and hence necessitates the administration of high gemcitabine dose during chemotherapy. Photodynamic therapy (PDT), a more selective and minimally invasive treatment, may be used synergistically with gemcitabine to reduce the doses utilized and dose-related side effects. This study reports the synergistic use of Re(I) bisquinolinyl complex, a transition metal complex photosensitizer with gemcitabine against PDAC. Re(I) bisquinolinyl complex was found to act synergistically with gemcitabine against PDAC in vitro at various ratios. With the aim to enhance cellular uptake and therapeutic efficiency, the Re(I) bisquinolinyl complex and gemcitabine were encapsulated into liquid crystalline nanoparticles (LCNPs) system. The formulations were found to produce homogeneous drug-loaded LCNPs (average size: 159-173 nm, zeta potential +1.06 to -10 mV). Around 70% of gemcitabine and 90% of the Re(I) bisquinolinyl complex were found to be entrapped efficiently in the formulated LCNPs. The release rate of gemcitabine or/and the Re(I) bisquinolinyl complex loaded into LCNPs was evaluated in vitro, and the hydrophilic gemcitabine was released at a faster rate than the lipophilic Re(I) complex. LCNPs loaded with gemcitabine and Re(I) bisquinolinyl complex in a 1:1 ratio illustrated the best anti-cancer activity among the LCNP formulations (IC50 of BxPC3: 0.15 μM; IC50 of SW 1990: 0.76 μM) through apoptosis. The current findings suggest the potential use of transition metal-based photosensitizer as an adjunctive agent for gemcitabine-based chemotherapy against PDAC and the importance of nano-formulation in such application.
Collapse
Affiliation(s)
- Hui Shan Liew
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Chun-Wai Mai
- Centre for Cancer and Stem Cell Research, International Medical University, Kuala Lumpur, Malaysia.,School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Mohd Zulkefeli
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | | | - Lik Voon Kiew
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Lesley Jia Wei Pua
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Ling Wei Hii
- Centre for Cancer and Stem Cell Research, International Medical University, Kuala Lumpur, Malaysia.,School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Wei Meng Lim
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
| | - May Lee Low
- Centre for Cancer and Stem Cell Research, International Medical University, Kuala Lumpur, Malaysia.,School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
7
|
Rai ZL, Ranieri V, Palmer DH, Littler P, Ghaneh P, Gurusamy K, Manas D, Pizzo E, Psarelli EE, Gilmore R, Peddu P, Bartlett DC, de Liguori Carino N, Davidson BR. Treatment of unresectable locally advanced pancreatic cancer with percutaneous irreversible electroporation (IRE) following initial systemic chemotherapy (LAP-PIE) trial: study protocol for a feasibility randomised controlled trial. BMJ Open 2022; 12:e050166. [PMID: 35551086 PMCID: PMC9109032 DOI: 10.1136/bmjopen-2021-050166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 01/17/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Approximately 30% of patients with pancreas cancer have unresectable locally advanced disease, which is currently treated with systemic chemotherapy. A new treatment option of irreversible electroporation (IRE) has been investigated for these patients since 2005. Cohort studies suggest that IRE confers a survival advantage, but with associated, procedure-related complications. Selection bias may account for improved survival and there have been no prospective randomised trials evaluating the harms and benefits of therapy. The aim of this trial is to evaluate the feasibility of a randomised comparison of IRE therapy with chemotherapy versus chemotherapy alone in patients with locally advanced pancreatic cancer (LAPC). METHODS AND ANALYSIS Eligible patients with LAPC who have undergone first-line 5-FluoroUracil, Leucovorin, Irinotecan and Oxaliplatin chemotherapy will be randomised to receive either a single session of IRE followed by (if indicated) further chemotherapy or to chemotherapy alone (standard of care). Fifty patients from up to seven specialist pancreas centres in the UK will be recruited over a period of 15 months. Trial follow-up will be 12 months. The primary outcome measure is ability to recruit. Secondary objectives include practicality and technical success of treatment, acceptability of treatment to patients and clinicians and safety of treatment. A qualitative study has been incorporated to evaluate the patient and clinician perspective of the locally advanced pancreatic cancer with percutaneous irreversible electroporation trial. It is likely that the data obtained will guide the structure, the primary outcome measure, the power and the duration of a subsequent multicentre randomised controlled trial aimed at establishing the clinical efficiency of pancreas IRE therapy. Indicative procedure-related costings will be collected in this feasibility trial, which will inform the cost evaluation in the subsequent study on efficiency. ETHICS AND DISSEMINATION The protocol has received approval by London-Brent Research Ethics Committee reference number 21/LO/0077.Results will be analysed following completion of trial recruitment and follow-up. Results will be presented to international conferences with an interest in oncology, hepatopancreaticobiliary surgery and interventional radiology and be published in a peer-reviewed journal. TRIAL REGISTRATION NUMBER ISRCTN14986389.
Collapse
Affiliation(s)
- Zainab L Rai
- Division of Surgery and Interventional Science, University College London, London, UK
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences (WEISS), London, UK
| | - Veronica Ranieri
- Research & Development, Tavistock and Portman NHS Foundation Trust, London, UK
- Science & Technology Studies, University College London, London, UK
| | - Daniel H Palmer
- Hepato-Pancreatco-Biliary Disease, University of Liverpool, Liverpool, Merseyside, UK
| | - Peter Littler
- Hepato-Pancreatico-Biliary Surgery, Freeman Hospital, Newcastle Upon Tyne, UK
| | - Pauleh Ghaneh
- Hepato-Pancreatco-Biliary Disease, University of Liverpool, Liverpool, Merseyside, UK
| | - Kurinchi Gurusamy
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Derek Manas
- Hepato-Pancreatico-Biliary Surgery, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle Upon Tyne, UK
| | - Elena Pizzo
- Applied Health Research, University College London, London, UK
| | | | - Roopinder Gilmore
- Division of Surgery and Interventional Science, University College London, London, UK
- Oncology, Royal Free London NHS Foundation Trust, London, UK
| | - Praveen Peddu
- Department of Radiology, King's College Hospital NHS Trust, London, UK
| | - David C Bartlett
- Hepato-Pancreatico-Biliary Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | | | | |
Collapse
|
8
|
Razaee ZS, Cook-Wiens G, Tighiouart M. A nonparametric Bayesian method for dose finding in drug combinations cancer trials. Stat Med 2022; 41:1059-1080. [PMID: 35075652 PMCID: PMC8881404 DOI: 10.1002/sim.9316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 10/18/2021] [Accepted: 12/19/2021] [Indexed: 11/11/2022]
Abstract
We propose an adaptive design for early-phase drug-combination cancer trials with the goal of estimating the maximum tolerated dose (MTD). A nonparametric Bayesian model, using beta priors truncated to the set of partially ordered dose combinations, is used to describe the probability of dose limiting toxicity (DLT). Dose allocation between successive cohorts of patients is estimated using a modified continual reassessment scheme. The updated probabilities of DLT are calculated with a Gibbs sampler that employs a weighting mechanism to calibrate the influence of data vs the prior. At the end of the trial, we recommend one or more dose combinations as the MTD based on our proposed algorithm. We apply our method to a Phase I clinical trial of CB-839 and Gemcitabine that motivated this nonparametric design. The design operating characteristics indicate that our method is comparable with existing methods.
Collapse
Affiliation(s)
- Zahra S Razaee
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Galen Cook-Wiens
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mourad Tighiouart
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
9
|
Hewitt DB, Nissen N, Hatoum H, Musher B, Seng J, Coveler AL, Al-Rajabi R, Yeo CJ, Leiby B, Banks J, Balducci L, Vaccaro G, LoConte N, George TJ, Brenner W, Elquza E, Vahanian N, Rossi G, Kennedy E, Link C, Lavu H. A Phase 3 Randomized Clinical Trial of Chemotherapy With or Without Algenpantucel-L (HyperAcute-Pancreas) Immunotherapy in Subjects With Borderline Resectable or Locally Advanced Unresectable Pancreatic Cancer. Ann Surg 2022; 275:45-53. [PMID: 33630475 DOI: 10.1097/sla.0000000000004669] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To compare the efficacy and safety of algenpantucel-L [HyperAcute-Pancreas algenpantucel-L (HAPa); IND# 12311] immunotherapy combined with standard of care (SOC) chemotherapy and chemoradiation to SOC chemotherapy and chemoradiation therapy alone in patients with borderline resectable or locally advanced pancreatic ductal adenocarcinoma (PDAC). SUMMARY BACKGROUND DATA To date, immunotherapy has not been shown to benefit patients with borderline resectable or locally advanced unresectable PDAC. HAPa is a cancer vaccine consisting of allogeneic pancreatic cancer cells engineered to express the murine α(1,3)GT gene. METHODS A multicenter, phase 3, open label, randomized (1:1) trial of patients with borderline resectable or locally advanced unresectable PDAC. Patients received neoadjuvant SOC chemotherapy (FOLFIRINOX or gemcitabine/nab-paclitaxel) followed by chemoradiation (standard group) or the same standard neoadjuvant regimen combined with HAPa immunotherapy (experimental group). The primary outcome was overall survival. RESULTS Between May 2013 and December 2015, 303 patients were randomized from 32 sites. Median (interquartile range) overall survival was 14.9 (12.2-17.8) months in the standard group (N = 158) and 14.3 (12.6-16.3) months in the experimental group (N = 145) [hazard ratio (HR) 1.02, 95% confidence intervals 0.66-1.58; P = 0.98]. Median progression-free survival was 13.4 months in the standard group and 12.4 months in the experimental group (HR 1.33, 95% confidence intervals 0.72-1.78; P = 0.59). Grade 3 or higher adverse events occurred in 105 of 140 patients (75%) in the standard group and in 115 of 142 patients (81%) in the experimental group (P > 0.05). CONCLUSIONS Algenpantucel-L immunotherapy did not improve survival in patients with borderline resectable or locally advanced unresectable PDAC receiving SOC neoadjuvant chemotherapy and chemoradiation. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01836432.
Collapse
Affiliation(s)
- Daniel Brock Hewitt
- Thomas Jefferson University Hospital and The Jefferson Pancreas, Biliary and Related Cancer Center, Philadelphia, Pennsylvania
| | | | | | | | - John Seng
- Virginia Piper Cancer Institute, Minneapolis, Minnesota
| | - Andrew L Coveler
- University of Washington-Seattle Cancer Care, Seattle, Washington
| | | | - Charles J Yeo
- Thomas Jefferson University Hospital and The Jefferson Pancreas, Biliary and Related Cancer Center, Philadelphia, Pennsylvania
| | - Benjamin Leiby
- Thomas Jefferson University Hospital and The Jefferson Pancreas, Biliary and Related Cancer Center, Philadelphia, Pennsylvania
| | - Joshua Banks
- Thomas Jefferson University Hospital and The Jefferson Pancreas, Biliary and Related Cancer Center, Philadelphia, Pennsylvania
| | | | - Gina Vaccaro
- Oregon Health and Science University, Portland, Oregon
| | | | | | | | - Emad Elquza
- University of California at Irvine, Irvine, California
| | | | | | - Eugene Kennedy
- NewLink Genetics Corporation, Ames, Iowa
- Lumos Pharma Inc, Ames, Iowa
| | | | - Harish Lavu
- Thomas Jefferson University Hospital and The Jefferson Pancreas, Biliary and Related Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Hani U, Osmani RAM, Siddiqua A, Wahab S, Batool S, Ather H, Sheraba N, Alqahtani A. A systematic study of novel drug delivery mechanisms and treatment strategies for pancreatic cancer. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Cazes A, Betancourt O, Esparza E, Mose ES, Jaquish D, Wong E, Wascher AA, Tiriac H, Gymnopoulos M, Lowy AM. A MET Targeting Antibody-Drug Conjugate Overcomes Gemcitabine Resistance in Pancreatic Cancer. Clin Cancer Res 2021; 27:2100-2110. [PMID: 33451980 DOI: 10.1158/1078-0432.ccr-20-3210] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/16/2020] [Accepted: 01/08/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Pancreatic cancer is an aggressive disease associated with a poor 5-year overall survival. Most patients are ineligible for surgery due to late diagnosis and are treated primarily with chemotherapy with very limited success. Pancreatic cancer is relatively insensitive to chemotherapy due to multiple factors, including reduced bioavailability of drugs to tumor cells. One strategy to improve drug efficacy with reduced toxicity is the development of antibody-drug conjugates (ADC), which have now been used successfully to treat both solid and liquid tumors. Here, we evaluate the efficacy of TR1801-ADC, a newly developed ADC composed of a MET antibody conjugated to the highly potent pyrrolobenzodiazepine toxin-linker, tesirine. EXPERIMENTAL DESIGN We first evaluated MET expression and subcellular localization in pancreatic cancer cell lines, human tumors, and patient-derived xenografts (PDX). We then tested TR1801-ADC efficacy in vitro in pancreatic cancer cell lines. Preclinical evaluation of TR1801-ADC efficacy was conducted on PDXs selected on the basis of their MET expression level. RESULTS We show that MET is highly expressed and located at the plasma membrane of pancreatic cancer cells. We found that TR1801-ADC induces a specific cytotoxicity in pancreatic cancer cell lines and a profound tumor growth inhibition, even in a gemcitabine-resistant tumor. We also noted synergism between TR1801-ADC and gemcitabine in vitro and an improved response to the combination in vivo. CONCLUSIONS Together, these results suggest the promise of agents such as TR1801-ADC as a novel approach to the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Alex Cazes
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California San Diego, La Jolla, California
| | | | - Edgar Esparza
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Evangeline S Mose
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Dawn Jaquish
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Eric Wong
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Alexis A Wascher
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Hervé Tiriac
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California San Diego, La Jolla, California
| | | | - Andrew M Lowy
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California San Diego, La Jolla, California.
| |
Collapse
|
12
|
Gan LL, Hii LW, Wong SF, Leong CO, Mai CW. Molecular Mechanisms and Potential Therapeutic Reversal of Pancreatic Cancer-Induced Immune Evasion. Cancers (Basel) 2020; 12:cancers12071872. [PMID: 32664564 PMCID: PMC7408947 DOI: 10.3390/cancers12071872] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer ranks high among the causes of cancer-related mortality. The prognosis of this grim condition has not improved significantly over the past 50 years, despite advancement in imaging techniques, cancer genetics and treatment modalities. Due to the relative difficulty in the early detection of pancreatic tumors, as low as 20% of patients are eligible for potentially curative surgery; moreover, chemotherapy and radiotherapy (RT) do not confer a great benefit in the overall survival of the patients. Currently, emerging developments in immunotherapy have yet to bring a significant clinical advantage among pancreatic cancer patients. In fact, pancreatic tumor-driven immune evasion possesses one of the greatest challenges leading to immunotherapeutic resistance. Most of the immune escape pathways are innate, while poor priming of hosts' immune response and immunoediting constitute the adaptive immunosuppressive machinery. In this review, we extensively discuss the pathway perturbations undermining the anti-tumor immunity specific to pancreatic cancer. We also explore feasible up-and-coming therapeutic strategies that may restore immunity and address therapeutic resistance, bringing hope to eliminate the status quo in pancreatic cancer prognosis.
Collapse
Affiliation(s)
- Li-Lian Gan
- School of Postgraduate Study, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (L.-L.G.); (L.-W.H.)
| | - Ling-Wei Hii
- School of Postgraduate Study, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (L.-L.G.); (L.-W.H.)
- School of Pharmacy, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
| | - Shew-Fung Wong
- School of Medicine, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- Centre for Environmental and Population Health, Institute for Research, Development and Innovation (IRDI), International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Chee-Onn Leong
- School of Pharmacy, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- Centre for Cancer and Stem Cells Research, Institute for Research, Development and Innovation (IRDI), International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Chun-Wai Mai
- School of Pharmacy, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- Centre for Cancer and Stem Cells Research, Institute for Research, Development and Innovation (IRDI), International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia
- Correspondence: ; Tel.: +60-3-2731-7596
| |
Collapse
|
13
|
Bakshi HA, Zoubi MSA, Faruck HL, Aljabali AAA, Rabi FA, Hafiz AA, Al-Batanyeh KM, Al-Trad B, Ansari P, Nasef MM, Charbe NB, Satija S, Mehta M, Mishra V, Gupta G, Abobaker S, Negi P, Azzouz IM, Dardouri AAK, Dureja H, Prasher P, Chellappan DK, Dua K, Silva MWD, Tanani ME, McCarron PA, M. Tambuwala M. Dietary Crocin is Protective in Pancreatic Cancer while Reducing Radiation-Induced Hepatic Oxidative Damage. Nutrients 2020; 12:nu12061901. [PMID: 32604971 PMCID: PMC7353213 DOI: 10.3390/nu12061901] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer is one of the fatal causes of global cancer-related deaths. Although surgery and chemotherapy are standard treatment options, post-treatment outcomes often end in a poor prognosis. In the present study, we investigated anti-pancreatic cancer and amelioration of radiation-induced oxidative damage by crocin. Crocin is a carotenoid isolated from the dietary herb saffron, a prospect for novel leads as an anti-cancer agent. Crocin significantly reduced cell viability of BXPC3 and Capan-2 by triggering caspase signaling via the downregulation of Bcl-2. It modulated the expression of cell cycle signaling proteins P53, P21, P27, CDK2, c-MYC, Cyt-c and P38. Concomitantly, crocin treatment-induced apoptosis by inducing the release of cytochrome c from mitochondria to cytosol. Microarray analysis of the expression signature of genes induced by crocin showed a substantial number of genes involved in cell signaling pathways and checkpoints (723) are significantly affected by crocin. In mice bearing pancreatic tumors, crocin significantly reduced tumor burden without a change in body weight. Additionally, it showed significant protection against radiation-induced hepatic oxidative damage, reduced the levels of hepatic toxicity and preserved liver morphology. These findings indicate that crocin has a potential role in the treatment, prevention and management of pancreatic cancer.
Collapse
Affiliation(s)
- Hamid A. Bakshi
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; (M.W.D.S.); (P.A.M.)
- Correspondence: or (H.A.B.); (H.L.F.); (M.M.T.)
| | - Mazhar S Al Zoubi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid 566, Jordan;
| | - Hakkim L. Faruck
- Department of Mathematics and Sciences, College of Arts and Applied Sciences, Dhofar University, Salalah 211, Oman
- Correspondence: or (H.A.B.); (H.L.F.); (M.M.T.)
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 566, Jordan;
| | - Firas A. Rabi
- Department of Clinical Sciences, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan;
| | - Amin A. Hafiz
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21421, Saudi Arabia;
| | - Khalid M Al-Batanyeh
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 566, Jordan; (K.M.A.-B.); (B.A.-T.)
| | - Bahaa Al-Trad
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 566, Jordan; (K.M.A.-B.); (B.A.-T.)
| | - Prawej Ansari
- School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK;
| | - Mohamed M. Nasef
- Department of Pharmacy and Biomedical Sciences, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield HD13DH, UK;
| | - Nitin B. Charbe
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins, Santiago 340, Región Metropolitana, Chile;
| | - Saurabh Satija
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; (S.S.); (M.M.); (V.M.)
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Meenu Mehta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; (S.S.); (M.M.); (V.M.)
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; (S.S.); (M.M.); (V.M.)
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, Rajasthan 302017, India;
| | - Salem Abobaker
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow, Klinikum Charite-Universitatmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University, Bajhol, Sultanpur, Solan, Himachal Pradesh 173229, India;
| | - Ibrahim M. Azzouz
- Department of Dermatology, Venerology, and Allergology, Charite-Universitatsmedizin Berlin, Corporate Member of Freie Universitat Berlin, Chariteplatz1, 10117 Berlin, Germany;
| | - Ashref Ali K Dardouri
- Department of Forensic Science, School of Applied Sciences, University of Huddersfield, Huddersfield HD13DH, UK;
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana 124001, India;
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun 248007, India;
| | - Dinesh K. Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia;
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia;
- School of Pharmaceutical Sciences, Shoolini University, Bajhol, Sultanpur, Solan, Himachal Pradesh 173229, India;
| | - Mateus Webba Da Silva
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; (M.W.D.S.); (P.A.M.)
| | - Mohamed El Tanani
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan;
| | - Paul A. McCarron
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; (M.W.D.S.); (P.A.M.)
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; (M.W.D.S.); (P.A.M.)
- Correspondence: or (H.A.B.); (H.L.F.); (M.M.T.)
| |
Collapse
|
14
|
Wolfe AR, Prabhakar D, Yildiz VO, Cloyd JM, Dillhoff M, Abushahin L, Alexandra Diaz D, Miller ED, Chen W, Frankel WL, Noonan A, Williams TM. Neoadjuvant-modified FOLFIRINOX vs nab-paclitaxel plus gemcitabine for borderline resectable or locally advanced pancreatic cancer patients who achieved surgical resection. Cancer Med 2020; 9:4711-4723. [PMID: 32415696 PMCID: PMC7333854 DOI: 10.1002/cam4.3075] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 04/02/2020] [Accepted: 04/05/2020] [Indexed: 12/11/2022] Open
Abstract
We conducted an institutional study to compare the clinical and pathological efficacy between the neoadjuvant therapy (NAT)‐modified FOLFIRINOX (mFOLF) vs nanoparticle albumin–bound paclitaxel plus gemcitabine (nab‐P/G) for borderline resectable pancreatic cancer (BRPC) and locally advanced pancreatic cancer (LAPC) patients who completed resection. The study retrospectively enrolled patients with pathologically confirmed BRPC or LAPC from 2010 to 2018 at our institution. The survival rates were determined by the Kaplan‐Meier method and log‐rank test was used to test differences. Cox's proportional hazard model was used to assess survival with respect to covariates. Seventy‐two patients who completed at least two cycles of neoadjuvant chemotherapy and surgical resection were included, with 52 (72.2%) patients receiving mFOLF and 20 (27.8%) receiving nab‐P/G. Patients treated with mFOLF had statistically higher rates of RECIST 1.1 partial or complete response (16/52 vs 1/20, P = .028). Additionally, mFOLF patients had greater pathological tumor size reduction, fewer positive lymph nodes, and higher treatment response grade compared to the nab‐P/G patients (all P < .05). The median overall survival was 33.3 months vs 27.1 months (P = .105), and distant metastasis‒free survival (DMFS) was 21.3 months vs 14.6 months (P = .042) in the mFOLF vs nab‐P/G groups, respectively. On multivariate analysis, mFOLF (hazard ratio, 0.428; 95% confidence interval [CI], 0.186‐0.987) and abnormal postoperative CA 19‐9 (hazard ratio, 2.47; 95% CI, 1.06‐5.76) were associated with DMFS. Among patients with BRPC and LAPC who complete surgical resection, neoadjuvant mFOLF was associated with improved pathological and clinical outcomes compared with nab‐P/G.
Collapse
Affiliation(s)
- Adam R Wolfe
- Department of Radiation Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Dhivya Prabhakar
- Department of Medical Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Vedat O Yildiz
- Department of Biomedical Informatics, Ohio State College of Medicine, Columbus, OH, USA
| | - Jordan M Cloyd
- Department of Surgical Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Mary Dillhoff
- Department of Surgical Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Laith Abushahin
- Department of Medical Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Dayssy Alexandra Diaz
- Department of Radiation Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Eric D Miller
- Department of Radiation Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Wei Chen
- Department of Pathology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Wendy L Frankel
- Department of Pathology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Anne Noonan
- Department of Medical Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Terence M Williams
- Department of Radiation Oncology, Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
15
|
Urrutia G, Salmonson A, Toro-Zapata J, de Assuncao TM, Mathison A, Dusetti N, Iovanna J, Urrutia R, Lomberk G. Combined Targeting of G9a and Checkpoint Kinase 1 Synergistically Inhibits Pancreatic Cancer Cell Growth by Replication Fork Collapse. Mol Cancer Res 2019; 18:448-462. [PMID: 31822519 DOI: 10.1158/1541-7786.mcr-19-0490] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/31/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022]
Abstract
Because of its dismal outcome, pancreatic ductal adenocarcinoma (PDAC) remains a therapeutic challenge making the testing of new pharmacologic tools a goal of paramount importance. Here, we developed a rational approach for inhibiting PDAC growth based on leveraging cell-cycle arrest of malignant cells at a phase that shows increased sensitivity to distinct epigenomic inhibitors. Specifically, we simultaneously inhibited checkpoint kinase 1 (Chk1) by prexasertib and the G9a histone methyltransferase with BRD4770, thereby targeting two key pathways for replication fork stability. Methodologically, the antitumor effects and molecular mechanisms of the combination were assessed by an extensive battery of assays, utilizing cell lines and patient-derived cells as well as 3D spheroids and xenografts. We find that the prexasertib-BRD4770 combination displays a synergistic effect on replication-associated phenomena, including cell growth, DNA synthesis, cell-cycle progression at S phase, and DNA damage signaling, ultimately leading to a highly efficient induction of cell death. Moreover, cellular and molecular data reveal that the synergistic effect of these pathways can be explained, at least in large part, by the convergence of both Chk1 and G9a functions at the level of the ATR-RPA-checkpoint pathway, which is operational during replication stress. Thus, targeting the epigenetic regulator G9a, which is necessary for replication fork stability, combined with inhibition of the DNA damage checkpoint, offers a novel approach for controlling PDAC growth through replication catastrophe. IMPLICATIONS: This study offers an improved, context-dependent, paradigm for the use of epigenomic inhibitors and provides mechanistic insight into their potential therapeutic use against PDAC.
Collapse
Affiliation(s)
- Guillermo Urrutia
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ann Salmonson
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jorge Toro-Zapata
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Thiago M de Assuncao
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin.,Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Angela Mathison
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin.,Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nelson Dusetti
- 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, Marseille, France
| | - 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, Marseille, France
| | - Raul Urrutia
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin.,Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Gwen Lomberk
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin. .,Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
16
|
Rezaee M, Wang J, Razavi M, Ren G, Zheng F, Hussein A, Ullah M, Thakor AS. A Study Comparing the Effects of Targeted Intra-Arterial and Systemic Chemotherapy in an Orthotopic Mouse Model of Pancreatic Cancer. Sci Rep 2019; 9:15929. [PMID: 31685925 PMCID: PMC6828954 DOI: 10.1038/s41598-019-52490-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 10/18/2019] [Indexed: 02/06/2023] Open
Abstract
Systemic chemotherapy is the first line treatment for patients with unresectable pancreatic cancer, however, insufficient drug delivery to the pancreas is a major problem resulting in poor outcomes. We evaluated the therapeutic effects of targeted intra-arterial (IA) delivery of gemcitabine directly into the pancreas in an orthotopic mouse model of pancreatic cancer. Nude mice with orthotopic pancreatic tumors were randomly assigned into 3 groups receiving gemcitabine: systemic intravenous (IV) injection (low: 0.3 mg/kg and high: 100 mg/kg) and direct IA injection (0.3 mg/kg). Treatments were administered weekly for 2 weeks. IA treatment resulted in a significantly greater reduction in tumor growth compared to low IV treatment. To achieve a comparable reduction in tumor growth as seen with IA treatment, gemcitabine had to be given IV at over 300x the dose (high IV treatment) which was associated with some toxicity. After 2 weeks, tumor samples from animals treated with IA gemcitabine had significantly lower residual cancer cells, higher cellular necrosis and evidence of increased apoptosis when compared to animals treated with low IV gemcitabine. Our study shows targeted IA injection of gemcitabine directly into the pancreas, via its arterial blood supply, has a superior therapeutic effect in reducing tumor growth compared to the same concentration administered by conventional systemic injection.
Collapse
MESH Headings
- Administration, Intravenous
- Animals
- Antimetabolites, Antineoplastic/adverse effects
- Antimetabolites, Antineoplastic/therapeutic use
- Cell Line, Tumor
- Deoxycytidine/adverse effects
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/therapeutic use
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Female
- Humans
- Infusions, Intra-Arterial
- Male
- Mice
- Mice, Nude
- Neoplasm, Residual
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Transplantation, Heterologous
- Gemcitabine
Collapse
Affiliation(s)
- Melika Rezaee
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
- Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois, 60064, USA
| | - Jing Wang
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Gang Ren
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Fengyan Zheng
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Ahmed Hussein
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA.
| |
Collapse
|
17
|
Intra-arterial infusion chemotherapy versus isolated upper abdominal perfusion for advanced pancreatic cancer: a retrospective cohort study on 454 patients. J Cancer Res Clin Oncol 2019; 145:2855-2862. [PMID: 31506738 PMCID: PMC6800855 DOI: 10.1007/s00432-019-03019-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/03/2019] [Indexed: 02/08/2023]
Abstract
Purpose The treatment of pancreatic carcinoma remains a challenge as prognosis is poor, even if confined to a single anatomical region. A regional treatment of pancreatic cancer with high drug concentrations at the tumor site may increase response behaviour. Intra-arterial administration of drugs generates homogenous drug distribution throughout the entire tumor volume. Methods We report on treatment outcome of 454 patients with advanced pancreatic carcinoma (WHO stage III: 174 patients, WHO stage IV: 280 patients). Patients have been separated to two different treatment protocols. The first group (n = 233 patients) has been treated via angiographically placed celiac axis catheters. The second group (n = 221 patients) had upper abdominal perfusion (UAP) with stopflow balloon catheters in aorta and vena cava. Both groups have been treated with a combination of cisplatin, adriamycin and mitomycin. Results For stage III pancreatic cancer, median survival rates of 8 and 12 months were reached with IA and UAP treatment, respectively. For stage IV pancreatic cancer, median survival rates of 7 and 8.5 months were reached with IA and UAP treatment, respectively. Resolution of ascites has been reached in all cases by UAP treatment. Toxicity was generally mild, WHO grade I or II, toxicity grade III or IV was only noted in patients with severe systemic pretreatment. The techniques, survival data and detailed results are demonstrated. Conclusions Responsiveness of pancreatic cancer to regional chemotherapy is drug exposure dependent. The isolated perfusion procedure is superior to intra-arterial infusion in survival times. Electronic supplementary material The online version of this article (10.1007/s00432-019-03019-6) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Woo W, Carey ET, Choi M. Spotlight on liposomal irinotecan for metastatic pancreatic cancer: patient selection and perspectives. Onco Targets Ther 2019; 12:1455-1463. [PMID: 30863113 PMCID: PMC6391121 DOI: 10.2147/ott.s167590] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is a highly lethal disease, where the mortality closely matches increasing incidence. Pancreatic ductal adenocarcinoma (PDAC) is the most common histologic type that tends to metastasize early in tumor progression. For metastatic PDAC, gemcitabine had been the mainstay treatment for the past three decades. The treatment landscape has changed since 2010, and current first-line chemotherapy includes triplet drugs like FOLFIRINOX (folinic acid, 5-fluorouracil, irinotecan, and oxaliplatin), and doublet agents like nab-paclitaxel and gemcitabine. Nanoliposomal encapsulated irinotecan (nal-IRI) was developed as a novel formulation to improve drug delivery, effectiveness, and limit toxicities. Nal-IRI, in combination with leucovorin-modulated fluorouracil (5-FU/LV), was found in a large randomized phase III clinical trial (NAPOLI-1) to significantly improve overall survival in patients who progressed on gemcitabine-based therapy. This review will focus on the value of using nal-IRI, toxicities, recent clinical experiences, and tools to improve patient outcomes in this setting.
Collapse
Affiliation(s)
- Wonhee Woo
- Division of Hematology/Oncology, Department of Medicine, Stony Brook University, Stony Brook, NY, USA,
| | - Edward T Carey
- Division of Hematology/Oncology, Department of Medicine, Stony Brook University, Stony Brook, NY, USA,
| | - Minsig Choi
- Division of Hematology/Oncology, Department of Medicine, Stony Brook University, Stony Brook, NY, USA,
| |
Collapse
|
19
|
Abstract
OBJECTIVES The benefit-risk balance of 5-fluorouracil, oxaliplatin, irinotecan, and leucovorin versus gemcitabine assessed using generalized pairwise comparison was strongly positive. We sought to assess the benefit-risk balance of nab-paclitaxel plus gemcitabine using the data of the MPACT trial, as it is an alternative to 5-fluorouracil, oxaliplatin, irinotecan, and leucovorin. METHODS This statistical method allows for the simultaneous analysis of several prioritized outcomes. The first priority outcome was survival time (overall survival). The second priority outcome was toxicity. The overall treatment effect was quantified using the overall net benefit. Multiple sensitivity analyses were performed to assess the consistency of the results according to possible patients' preferences. RESULTS In this trial, 861 patients received nab-paclitaxel plus gemcitabine or gemcitabine alone. The overall net benefit favored strongly and significantly the combination group. When only large survival differences were considered clinically relevant, the net benefit was not in favor of the combination group. CONCLUSIONS The overall net benefit is a clinically intuitive way of comparing patients with respect to all important efficacy and toxicity outcomes. The nab-paclitaxel plus gemcitabine combination has a positive benefit-risk balance, but it might not be suitable for patients who would consider losing several months of survival to avoid a significant toxic event.
Collapse
|
20
|
Liu H, Sun M, Liu Z, Kong C, Kong W, Ye J, Gong J, Huang DCS, Qian F. KRAS-enhanced macropinocytosis and reduced FcRn-mediated recycling sensitize pancreatic cancer to albumin-conjugated drugs. J Control Release 2019; 296:40-53. [PMID: 30653981 DOI: 10.1016/j.jconrel.2019.01.014] [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: 10/27/2018] [Revised: 01/09/2019] [Accepted: 01/14/2019] [Indexed: 11/26/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a dominantly (~95%) KRAS-mutant cancer that has extremely poor prognosis, in part this is due to its strong intrinsic resistance towards almost all therapeutic agents. PDAC relies heavily on KRAS-transformed metabolism, including enhanced macropinocytosis and catabolism of extracellular albumin, to maintain its proliferation and progression. However, it has yet to be validated that whether such transformed metabolism could be exploited for the drug delivery to open therapeutic windows of cytotoxic agents in KRAS-mutant PDAC. In this study, we attempt to answer this question by focusing on the impact of two critical regulators of albumin catabolism, KRAS and the neonatal Fc receptor (FcRn), on the sensitivity of PDAC to doxorubicin (DOX, a model cytotoxic agent) and albumin-conjugated doxorubicin (DOX-ALB). Using cell lines and cell-derived xenografts with different KRAS genotypes and FcRn levels, we demonstrated that KRAS-enhanced macropinocytosis and reduced FcRn expression sensitize PDAC to DOX-ALB but not free DOX. In both in vitro and in vivo comparsion, the DOX-ALB demonstrated ~10 times enlarged therapeutic window compared with free DOX, in PDAC with KRAS mutation and reduced FcRn level, two events appear to occur simultaneously in the investigated PDAC. In summary, we conclude that albumin conjugation is an exploitable drug delivery strategy that significantly opens the therapeutic windows of otherwise undevelopable anti-cancer agents for KRAS-mutant PDAC therapy, and creates a new landscape for clinical evaluation and future translation of such compounds.
Collapse
Affiliation(s)
- Huiqin Liu
- School of Pharmaceutical Sciences and Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China
| | - Mengnan Sun
- School of Pharmaceutical Sciences and Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China
| | - Zhengsheng Liu
- School of Pharmaceutical Sciences and Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China
| | - Chao Kong
- School of Pharmaceutical Sciences and Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China
| | - Weijian Kong
- School of Pharmaceutical Sciences and Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China
| | - Junxiao Ye
- School of Pharmaceutical Sciences and Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China
| | - Jianan Gong
- Departments of Medical Biology, University of Melbourne, Australia; The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - David C S Huang
- Departments of Medical Biology, University of Melbourne, Australia; The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Feng Qian
- School of Pharmaceutical Sciences and Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
21
|
Prognostic factors for survival with nab-paclitaxel plus gemcitabine in metastatic pancreatic cancer in real-life practice: the ANICE-PaC study. BMC Cancer 2018; 18:1185. [PMID: 30497432 PMCID: PMC6267080 DOI: 10.1186/s12885-018-5101-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 11/16/2018] [Indexed: 12/28/2022] Open
Abstract
Background Treatment with nab-paclitaxel plus gemcitabine increases survival in patients with metastatic pancreatic cancer. However, the assessment of treatment efficacy and safety in non-selected patients in a real-life setting may provide useful information to support decision-making processes in routine practice. Methods Retrospective, multicenter study including patients with metastatic pancreatic cancer, who started first-line treatment with nab-paclitaxel plus gemcitabine between December 2013 and June 2015 according to routine clinical practice. In addition to describing the treatment pattern, overall survival (OS) and progression-free survival (PFS) were assessed for the total sample and the exploratory subgroups based on the treatment and patients’ clinical characteristics. Results All 210 eligible patients had a median age of 65.0 years (range 37–81). Metastatic pancreatic adenocarcinoma was recurrent in 46 (21.9%) patients and de novo in 164 (78.1%); 38 (18%) patients had a biliary stent. At baseline, 33 (18.1%) patients had an ECOG performance status ≥2. Patients received a median of four cycles of treatment (range 1–21), with a median duration of 3.5 months; 137 (65.2%) patients had a dose reduction of nab-paclitaxel and/or gemcitabine during treatment, and 33 (17.2%) discontinued treatment due to toxicity. Relative dose intensity (RDI) for nab-paclitaxel, gemcitabine, and the combined treatment was 66.7%. Median OS was 7.2 months (95% CI 6.0–8.5), and median PFS was 5.0 months (95% CI 4.3–5.9); 50 patients achieved either a partial or complete response (ORR 24.6%). OS was influenced by baseline ECOG PS, NLR and CA 19.9, but not by age ≥ 70 years and/or the presence of hepatobiliary stent or RDI < 85%. All included variables, computed as dichotomous, showed a significant contribution to the Cox regression model to build a nomogram for predicting survival in these patients: baseline ECOG 0–1 vs. 2–3 (p = 0.030), baseline NLR > 3 vs. ≤ 3 (p = 0.043), and baseline CA 19.9 > 37 U/mL vs. ≤37 U/mL (p = 0.004). Conclusions Nab-Paclitaxel plus gemcitabine remain effective in a real-life setting, despite the high burden of dose reductions and poorer performance of these patients. A nomogram to predict survival using baseline ECOG performance status, NLR and CA 19.9 is proposed.
Collapse
|
22
|
Hajatdoost L, Sedaghat K, Walker EJ, Thomas J, Kosari S. Chemotherapy in Pancreatic Cancer: A Systematic Review. ACTA ACUST UNITED AC 2018; 54:medicina54030048. [PMID: 30344279 PMCID: PMC6122094 DOI: 10.3390/medicina54030048] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 07/05/2018] [Indexed: 02/06/2023]
Abstract
Background and Aim: Pancreatic cancer is one of the most fatal cancers. Cytotoxic chemotherapy remains the mainstream treatment for unresectable pancreatic cancer. This systematic review evaluated and compared the overall survival (OS) and progression-free survival (PFS) outcomes obtained from recent phase 2 and 3 clinical trials of pancreatic cancer chemotherapy. Materials and methods: Thirty-two studies were included and compared based on chemotherapy agents or combinations used. Additionally, outcomes of first-line versus second-line chemotherapy in pancreatic cancer were compared. Results: In studies that investigated the treatments in adjuvant settings, the highest OS reported was for S-1 in patients, who received prior surgical resection (46.5 months). In neoadjuvant settings, the combination of gemcitabine, docetaxel, and capecitabine prior to the surgical resection had promising outcomes (OS of 32.5 months). In non-adjuvant settings, the highest OS reported was for the combination of temsirolimus plus bevacizumab (34.0 months). Amongst studies that investigated second-line treatment, the highest OS reported was for the combination of gemcitabine plus cisplatin (35.5 months), then temsirolimus plus bevacizumab (34.0 months). Conclusions: There is a need to develop further strategies besides chemotherapy to improve the outcomes in pancreatic cancer treatment. Future studies should consider surgical interventions, combination chemotherapy, and individualized second-line treatment based on the prior chemotherapy.
Collapse
Affiliation(s)
- Leva Hajatdoost
- Department of Pharmaceutical Sciences, Baha'i Institute for Higher Education (BIHE), Tehran 11369, Iran.
| | - Keyvan Sedaghat
- Department of Pharmaceutical Sciences, Baha'i Institute for Higher Education (BIHE), Tehran 11369, Iran.
| | - Erin J Walker
- Discipline of Pharmacy, Faculty of Health, University of Canberra, Bruce, Canberra 2617 ACT, Australia.
| | - Jackson Thomas
- Discipline of Pharmacy, Faculty of Health, University of Canberra, Bruce, Canberra 2617 ACT, Australia.
| | - Sam Kosari
- Discipline of Pharmacy, Faculty of Health, University of Canberra, Bruce, Canberra 2617 ACT, Australia.
| |
Collapse
|
23
|
Tahara J, Shimizu K, Otsuka N, Akao J, Takayama Y, Tokushige K. Gemcitabine plus nab-paclitaxel vs. FOLFIRINOX for patients with advanced pancreatic cancer. Cancer Chemother Pharmacol 2018; 82:245-250. [PMID: 29846765 DOI: 10.1007/s00280-018-3611-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/25/2018] [Indexed: 12/29/2022]
Abstract
PURPOSE New chemotherapies have become available for the treatment of advanced pancreatic cancer and have led to changes in its standard treatments. Since pancreatic cancer is becoming more common as a result of population aging, there is a need for diversification of chemotherapy. METHODS Between March 2014 and April 2017, FOLFIRINOX (FFX) or gemcitabine plus nab-paclitaxel (G-nab) was used as first-line therapy to treat 27 patients with locally advanced and metastatic pancreatic cancer at our hospital. In this study, we retrospectively evaluated their clinical characteristics, survival outcomes and adverse events. RESULTS Twelve of the 27 patients were treated with FFX, and the other 15 patients were treated with G-nab. The disease control rate was 86.7% in the G-nab group and 75% in the FFX group. Median OS time was 8.9 months in the FFX group and 11.8 months in the G-nab group. The 1-year survival rate was 46.6% in the G-nab group and 16.6% in the FFX group. The second-line treatment rate was 40% in the G-nab group and 66.7% in the FFX group. The grade 3-4 neutropenia rate was 20% in the G-nab group and 25% in the FFX group. No patients developed febrile neutropenia, or severe nausea, diarrhea, or anorexia. The peripheral sensory neuropathy rate was 73.3% in the G-nab group and 75% in the FFX group. CONCLUSIONS Although G-nab and FFX are effective treatments for advanced pancreatic cancer, the G-nab group had a higher 1-year survival rate, and G-nab can be more safely administered to older patients.
Collapse
Affiliation(s)
- Junko Tahara
- Department of Medicine, Institute of Gastroenterology, Tokyo Women's Medical University, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Kyoko Shimizu
- Department of Medicine, Institute of Gastroenterology, Tokyo Women's Medical University, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Nao Otsuka
- Department of Medicine, Institute of Gastroenterology, Tokyo Women's Medical University, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Junichi Akao
- Department of Medicine, Institute of Gastroenterology, Tokyo Women's Medical University, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Yukiko Takayama
- Department of Medicine, Institute of Gastroenterology, Tokyo Women's Medical University, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Katsutoshi Tokushige
- Department of Medicine, Institute of Gastroenterology, Tokyo Women's Medical University, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| |
Collapse
|
24
|
Kasten BB, Gangrade A, Kim H, Fan J, Ferrone S, Ferrone CR, Zinn KR, Buchsbaum DJ. 212Pb-labeled B7-H3-targeting antibody for pancreatic cancer therapy in mouse models. Nucl Med Biol 2017; 58:67-73. [PMID: 29413459 DOI: 10.1016/j.nucmedbio.2017.12.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/02/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022]
Abstract
INTRODUCTION We recently validated monoclonal antibody (mAb) 376.96 as an effective carrier for targeted α-particle radioimmunotherapy (RIT) with 212Pb in ovarian cancer mouse models. In this study, we tested the binding of radiolabeled mAb 376.96 to human pancreatic ductal adenocarcinoma (PDAC) cells and localization in xenografts in immune-deficient mice and evaluated 212Pb-labeled 376.96 (212Pb-376.96) for PDAC therapy. METHODS In vitro Scatchard assays assessed the specific binding of 212Pb-376.96 to human PDAC3 adherent differentiated cells and non-adherent cancer initiating cells (CICs) dissociated from tumorspheres. In vitro clonogenic assays were used to measure the proliferation of adherent PDAC3 cells and CIC-enriched tumorspheres treated with 212Pb-376.96 or the irrelevant isotype-matched 212Pb-F3-C25. Mice bearing patient derived pancreatic cancer Panc039 xenografts were i.v. injected with 0.17-0.70 MBq 212Pb-376.96 or isotype control 212Pb-F3-C25, and used for biodistribution and tumor growth inhibition studies. Mice bearing orthotopic PDAC3 xenografts were i.v. co-injected with 99mTc-376.96 and 125I-F3-C25 and used for biodistribution studies. RESULTS 212Pb-376.96 specifically bound to PDAC3 adherent and dissociated tumorsphere CICs; Kd values averaged 9.0 and 21.7 nM, respectively, with 104-105 binding sites/cell. 212Pb-376.96 inhibited the clonogenic survival of PDAC3 cells or CICs dissociated from tumorspheres 3-6 times more effectively than isotype-matched control 212Pb-F3-C25. Panc039 s.c. tumors showed significantly higher uptake of 212Pb-376.96 (14.0 ± 2.1% ID/g) compared to 212Pb-F3-C25 (6.5 ± 0.9% ID/g, p < .001) at 24 h after dosing. Orthotopic PDAC3 tumors showed significantly higher uptake of 99mTc-376.96 (6.4 ± 1.8% ID/g) compared to 125I-F3-C25 (3.9 ± 0.9% ID/g, p < .05) at 24 h after dosing. Panc039 tumor growth was significantly inhibited by 212Pb-376.96 compared to 212Pb-F3-C25 or non-treated control tumors (p < .05). CONCLUSION Our results provide evidence for the efficacy of B7-H3 targeted RIT against preclinical models of pancreatic ductal adenocarcinoma (PDAC) and support future studies with 212Pb-376.96 in combination with chemotherapy to potentiate efficacy against PDAC.
Collapse
Affiliation(s)
- Benjamin B Kasten
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States.
| | - Abhishek Gangrade
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Harrison Kim
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jinda Fan
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Kurt R Zinn
- Institute for Quantitative Health Science and Engineering, Department of Radiology, Michigan State University, East Lansing, MI, United States
| | - Donald J Buchsbaum
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
25
|
Liu P, Feng J, Sun M, Yuan W, Xiao R, Xiong J, Huang X, Xiong M, Chen W, Yu X, Sun Q, Zhao X, Zhang Q, Shao L. Synergistic effects of baicalein with gemcitabine or docetaxel on the proliferation, migration and apoptosis of pancreatic cancer cells. Int J Oncol 2017; 51:1878-1886. [DOI: 10.3892/ijo.2017.4153] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 09/18/2017] [Indexed: 11/06/2022] Open
|
26
|
Zarogoulidis P, Huang H, Bai C, Petridis D, Papadopoulou S, Faniadou E, Eleftheriadou E, Trakada G, Cristoforos K, Rapti A, Yarmus L, Kopman DF, Man YG, Hohenforst-Schmidt W. Nab-paclitaxel as First Line Treatment for NSCLC in Elderly Patients More Than 75 Years Old. J Cancer 2017; 8:1673-1678. [PMID: 28775787 PMCID: PMC5535723 DOI: 10.7150/jca.19463] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/03/2017] [Indexed: 12/30/2022] Open
Abstract
Introduction: Lung cancer is still the leading cause of cancer among cancer patients. Although there are novel therapies as second line treatment for NSCLC, there is an issue for elderly patients. Patients and Methods: We collected retrospectively data from 60 patients >75 years of age. Thirty of these patients received nab-paclitaxel and first line treatment and were compared to thirty patients that received only best supportive care. Results: The median life of patients at the date of disease progression, although increased by the administration of the drug (92 days versus 70) was not confirmed statistically significantly (Mann-Whitney test: W = 280, p = 0.138). The administration of drug seems to keep stable the biological condition of patients (McNemar's test: χ2 = 0.033, p = 0.99). Patients with chemotherapy the death rate was increased by 50% as compared to those with best supportive care (12 vs 8), the median life until the unfortunate event surpassed statistically significantly the latter (150 days of life as compared to 108, Mann-Whitney test: W = 57.5, p = 0.045). Discussion: Nab-paclitaxel as a monotherapy could be considered as a first line treatment option for patients > 75 years of age without any previous cardiological medical history when compared to best supportive care.
Collapse
Affiliation(s)
- Paul Zarogoulidis
- Pulmonary Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Haidong Huang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Chong Bai
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Dimitris Petridis
- Department of Food Technology, School of Food Technology and Nutrition, Alexander Technological Educational Institute, Thessaloniki, Greece
| | - Susana Papadopoulou
- Department of Nutrition and Dietetics, Alexander Technological Educational Institute, Thessaloniki, Greece
| | - Eleni Faniadou
- Pulmonary Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ellada Eleftheriadou
- Pulmonary Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia Trakada
- Division of Pulmonology, Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Alexandra Hospital, Athens, Greece
| | - Kosmidis Cristoforos
- General Surgery Department, European Interbalkan Medical Center, Thessaloniki, Greece
| | - Aggeliki Rapti
- Second Pulmonary Clinic, 'Sotiria' Chest Diseases Hospital, Athens, Greece
| | - Lonny Yarmus
- Division of Pulmonary and Critical Care Medicine, Sheikh Zayed Cardiovascular & Critical Care Tower, Baltimore, U.S.A
| | - David-Feller Kopman
- Division of Pulmonary and Critical Care Medicine, Sheikh Zayed Cardiovascular & Critical Care Tower, Baltimore, U.S.A
| | - Yan-Gao Man
- Research Laboratory and International Collaboration, Bon Secours Cancer Institute, VA, USA
| | | |
Collapse
|