1
|
Diab M. New Therapeutic Targets in RAS Wild-type Pancreatic Cancer. Curr Treat Options Oncol 2024; 25:1556-1562. [PMID: 39546212 DOI: 10.1007/s11864-024-01242-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 11/17/2024]
Abstract
OPINION STATEMENT The landscape of treatment of advanced PDAC is witnessing significant changes. This is in part due to the advent of molecular profiling, which has highlighted molecularly-distinct subsets of pts, especially those with KRAS wild-type disease. We now know that these pts harbor genomic alterations that not only serve as molecular drivers but also pose as therapeutically relevant markers. In the absence of strong evidence to support the use of targeted therapy in the front-line setting, we continue to offer chemotherapy for treatment-naïve pts. However, an argument can be made for the front-line use of targeted therapy in pts who are not fit for chemotherapy or who are not interested in it. The challenge is ensuring that molecular profiling is done in a timely fashion to prevent significant delays in therapy. In our practice, we offer molecular testing to all pts with a new diagnosis of advanced PDAC. We prefer the utility of targeted therapy in the second line and beyond for pts who have an actionable target, over the use of further chemotherapy, as targeted therapy appears to confer deep and durable responses and longer survival. For pts with MSI-H or MMRd disease, the use of immunotherapy is indicated, although it has to be noted that MSI-H/MMRd PDAC performed worse that other MSI-H/MMRd cancers treated with immunotherapy. Therefore, in the presence of MSI-H/MMRd and an additional actionable target, we prefer treating with targeted therapy and reserving immunotherapy for later lines. Pt preference has to be taken into consideration at all times though.
Collapse
Affiliation(s)
- Maria Diab
- Clinical Assistant Professor, Michigan State University/Henry Ford Health Sciences, 2800 W Grand Blvd, Detroit, MI, 48202, USA.
| |
Collapse
|
2
|
Jones L, Cunningham D, Starling N. HER-2 directed therapies across gastrointestinal tract cancers - A new frontier. Cancer Treat Rev 2024; 129:102789. [PMID: 38959629 DOI: 10.1016/j.ctrv.2024.102789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024]
Abstract
Gastrointestinal (GI) cancers are common and in the metastatic setting they have a poor prognosis. The current mainstay of treatment of GI cancers is chemotherapy; however, the biomarker-directed treatment landscape is evolving. HER-2 is overexpressed in a portion of GI cancers and is an emerging target for therapy, with recent FDA tumor agnostic approval for trastuzumab deruxtecan. Testing for HER-2 expression is not standardized across GI cancers, methodology requires further optimization and standardization as HER-2 targeted therapy emerges into the treatment landscape. There is established rationale for use of HER-2 targeted therapy in first line treatment of metastatic gastric cancer, and emerging evidence with variable benefit in bile duct, pancreatic and colorectal cancers.
Collapse
Affiliation(s)
- Lauren Jones
- Gastrointestinal and Lymphoma Unit, Royal Marsden NHS Foundation Trust, London, Sutton, UK
| | - David Cunningham
- Gastrointestinal and Lymphoma Unit, Royal Marsden NHS Foundation Trust, London, Sutton, UK
| | - Naureen Starling
- Gastrointestinal and Lymphoma Unit, Royal Marsden NHS Foundation Trust, London, Sutton, UK.
| |
Collapse
|
3
|
Bulle A, Liu P, Seehra K, Bansod S, Chen Y, Zahra K, Somani V, Khawar IA, Chen HP, Dodhiawala PB, Li L, Geng Y, Mo CK, Mahsl J, Ding L, Govindan R, Davies S, Mudd J, Hawkins WG, Fields RC, DeNardo DG, Knoerzer D, Held JM, Grierson PM, Wang-Gillam A, Ruzinova MB, Lim KH. Combined KRAS-MAPK pathway inhibitors and HER2-directed drug conjugate is efficacious in pancreatic cancer. Nat Commun 2024; 15:2503. [PMID: 38509064 PMCID: PMC10954758 DOI: 10.1038/s41467-024-46811-w] [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: 08/02/2023] [Accepted: 03/11/2024] [Indexed: 03/22/2024] Open
Abstract
Targeting the mitogen-activated protein kinase (MAPK) cascade in pancreatic ductal adenocarcinoma (PDAC) remains clinically unsuccessful. We aim to develop a MAPK inhibitor-based therapeutic combination with strong preclinical efficacy. Utilizing a reverse-phase protein array, we observe rapid phospho-activation of human epidermal growth factor receptor 2 (HER2) in PDAC cells upon pharmacological MAPK inhibition. Mechanistically, MAPK inhibitors lead to swift proteasomal degradation of dual-specificity phosphatase 6 (DUSP6). The carboxy terminus of HER2, containing a TEY motif also present in extracellular signal-regulated kinase 1/2 (ERK1/2), facilitates binding with DUSP6, enhancing its phosphatase activity to dephosphorylate HER2. In the presence of MAPK inhibitors, DUSP6 dissociates from the protective effect of the RING E3 ligase tripartite motif containing 21, resulting in its degradation. In PDAC patient-derived xenograft (PDX) models, combining ERK and HER inhibitors slows tumour growth and requires cytotoxic chemotherapy to achieve tumour regression. Alternatively, MAPK inhibitors with trastuzumab deruxtecan, an anti-HER2 antibody conjugated with cytotoxic chemotherapy, lead to sustained tumour regression in most tested PDXs without causing noticeable toxicity. Additionally, KRAS inhibitors also activate HER2, supporting testing the combination of KRAS inhibitors and trastuzumab deruxtecan in PDAC. This study identifies a rational and promising therapeutic combination for clinical testing in PDAC patients.
Collapse
Affiliation(s)
- Ashenafi Bulle
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Peng Liu
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kuljeet Seehra
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sapana Bansod
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yali Chen
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kiran Zahra
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Vikas Somani
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Iftikhar Ali Khawar
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hung-Po Chen
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Paarth B Dodhiawala
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lin Li
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yutong Geng
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chia-Kuei Mo
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jay Mahsl
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Li Ding
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ramaswamy Govindan
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sherri Davies
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jacqueline Mudd
- Section of Hepatobiliary Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - William G Hawkins
- Section of Hepatobiliary Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ryan C Fields
- Section of Hepatobiliary Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David G DeNardo
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | | | - Jason M Held
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Patrick M Grierson
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Andrea Wang-Gillam
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Marianna B Ruzinova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
4
|
Barzi A, Weipert CM, Espenschied CR, Raymond VM, Wang-Gillam A, Nezami MA, Gordon EJ, Mahadevan D, Mody K. ERBB2 (HER2) amplifications and co-occurring KRAS alterations in the circulating cell-free DNA of pancreatic ductal adenocarcinoma patients and response to HER2 inhibition. Front Oncol 2024; 14:1339302. [PMID: 38406801 PMCID: PMC10885695 DOI: 10.3389/fonc.2024.1339302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/16/2024] [Indexed: 02/27/2024] Open
Abstract
Purpose Despite accumulating data regarding the genomic landscape of pancreatic ductal adenocarcinoma (PDAC), olaparib is the only biomarker-driven FDA-approved targeted therapy with a PDAC-specific approval. Treating ERBB2(HER2)-amplified PDAC with anti-HER2 therapy has been reported with mixed results. Most pancreatic adenocarcinomas have KRAS alterations, which have been shown to be a marker of resistance to HER2-targeted therapies in other malignancies, though the impact of these alterations in pancreatic cancer is unknown. We describe two cases of ERBB2-amplified pancreatic cancer patients treated with anti-HER2 therapy and provide data on the frequency of ERBB2 amplifications and KRAS alterations identified by clinical circulating cell-free DNA testing. Methods De-identified molecular test results for all patients with pancreatic cancer who received clinical cell-free circulating DNA analysis (Guardant360) between 06/2014 and 01/2018 were analyzed. Cell-free circulating DNA analysis included next-generation sequencing of up to 73 genes, including select small insertion/deletions, copy number amplifications, and fusions. Results Of 1,791 patients with pancreatic adenocarcinoma, 36 (2.0%) had an ERBB2 amplification, 26 (72.2%) of whom had a KRAS alteration. Treatment data were available for seven patients. Two were treated with anti-HER2 therapy after their cell-free circulating DNA result, with both benefiting from therapy, including one with a durable response to trastuzumab and no KRAS alteration detected until progression. Conclusion Our case series illustrates that certain patients with ERBB2-amplified pancreatic adenocarcinoma may respond to anti-HER2 therapy and gain several months of prolonged survival. Our data suggests KRAS mutations as a possible mechanism of primary and acquired resistance to anti-HER2 therapy in pancreatic cancer. Additional studies are needed to clarify the role of KRAS in resistance to anti-HER2 therapy.
Collapse
Affiliation(s)
- Afsaneh Barzi
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, United States
| | | | | | | | - Andrea Wang-Gillam
- Division of Oncology, Siteman Cancer Center, St. Louis, MO, United States
| | | | - Eva J. Gordon
- Private Health Management, Inc., Los Angeles, CA, United States
| | - Daruka Mahadevan
- Division of Hematology and Oncology, Department of Medicine, University of Texas Health, San Antonio, San Antonio, TX, United States
| | - Kabir Mody
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
5
|
Lekan AA, Weiner LM. The Role of Chemokines in Orchestrating the Immune Response to Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:559. [PMID: 38339310 PMCID: PMC10854906 DOI: 10.3390/cancers16030559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Chemokines are small molecules that function as chemotactic factors which regulate the migration, infiltration, and accumulation of immune cells. Here, we comprehensively assess the structural and functional role of chemokines, examine the effects of chemokines that are present in the pancreatic ductal adenocarcinoma (PDAC) tumor microenvironment (TME), specifically those produced by cancer cells and stromal components, and evaluate their impact on immune cell trafficking, both in promoting and suppressing anti-tumor responses. We further explore the impact of chemokines on patient outcomes in PDAC and their role in the context of immunotherapy treatments, and review clinical trials that have targeted chemokine receptors and ligands in the treatment of PDAC. Lastly, we highlight potential strategies that can be utilized to harness chemokines in order to increase cytotoxic immune cell infiltration and the anti-tumor effects of immunotherapy.
Collapse
Affiliation(s)
| | - Louis M. Weiner
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC 20057, USA;
| |
Collapse
|
6
|
King DA, Smith AR, Pineda G, Nakano M, Michelini F, Goedegebuure SP, Thyparambil S, Liao WL, McCormick A, Ju J, Cioffi M, Zhang X, Hundal J, Griffith M, Grandori C, Pollastro M, Rosati R, Margossian A, Chatterjee P, Ainge T, Flory M, Ocampo P, Chen LM, Poultsides GA, Baron AD, Chang DT, Herman JM, Gillanders WE, Park H, Hoos WA, Nichols M, Fisher GA, Kuo CJ. Complete Remission of Widely Metastatic Human Epidermal Growth Factor Receptor 2-Amplified Pancreatic Adenocarcinoma After Precision Immune and Targeted Therapy With Description of Sequencing and Organoid Correlates. JCO Precis Oncol 2023; 7:e2100489. [PMID: 37079860 PMCID: PMC10309581 DOI: 10.1200/po.21.00489] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/10/2023] [Indexed: 04/22/2023] Open
Affiliation(s)
- Daniel A. King
- Northwell Health Cancer Institute and Feinstein Institute of Research, Lake Success, NY
| | | | | | - Michitaka Nakano
- Department of Medicine, Divisions of Hematology and Oncology, Stanford University School of Medicine, Stanford, CA
| | | | - S. Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine in St Louis, St Louis, MO
| | | | | | - Aaron McCormick
- Department of Medicine, Divisions of Hematology and Oncology, Stanford University School of Medicine, Stanford, CA
| | - Jihang Ju
- Department of Medicine, Divisions of Hematology and Oncology, Stanford University School of Medicine, Stanford, CA
| | | | - Xiuli Zhang
- Department of Surgery, Washington University School of Medicine in St Louis, St Louis, MO
| | - Jasreet Hundal
- Department of Surgery, Washington University School of Medicine in St Louis, St Louis, MO
| | - Malachi Griffith
- Department of Medicine, Washington University School of Medicine, St Louis, MO
| | | | | | | | | | | | | | - Marta Flory
- Department of Radiology, Stanford University, Stanford, CA
| | - Paolo Ocampo
- Personalized Healthcare, Genentech, Inc, South San Francisco, CA
| | - Lee-may Chen
- Department of Gynecologic Oncology, University of California at San Francisco, San Francisco, CA
| | - George A. Poultsides
- Department of Surgery, Section of Surgical Oncology, Stanford University, Stanford, CA
| | - Ari D. Baron
- Division of Hematology Oncology, California Pacific Medical Center, San Francisco, CA
| | - Daniel T. Chang
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford, CA
| | - Joseph M. Herman
- Department of Radiation Oncology and Northwell Health Cancer Institute, Lake Success, NY
| | - William E. Gillanders
- Department of Surgery, Washington University School of Medicine in St Louis, St Louis, MO
| | - Haeseong Park
- Department of Medicine, Division of Oncology, Washington University School of Medicine in St Louis
| | | | | | | | | |
Collapse
|
7
|
Pancreatic Cancer: Beyond Brca Mutations. J Pers Med 2022; 12:jpm12122076. [PMID: 36556296 PMCID: PMC9787452 DOI: 10.3390/jpm12122076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Pancreatic cancer is the fourth-leading cause of cancer-related deaths worldwide. The outcomes in patients with pancreatic cancer remain unsatisfactory. In the current review, we summarize the genetic and epigenetic architecture of metastatic pancreatic cancer beyond the BRCA mutations, focusing on the genetic alterations and the molecular pathology in pancreatic cancer. This review focuses on the molecular targets for the treatment of pancreatic cancer, with a correlation to future treatments. The potential approach addressed in this review may lead to the identification of a subset of patients with specific biological behaviors and treatment responses.
Collapse
|
8
|
Mao T, Zhang X, Xu H, Zhang X, Ge W, Li S, Ma J, Yue M, Xue S, Cui J, Wang L. HDACs/mTOR inhibitor synergizes with pyrotinib in HER2-positive pancreatic cancer through degradation of mutant P53. Cancer Cell Int 2022; 22:380. [PMID: 36457011 PMCID: PMC9714091 DOI: 10.1186/s12935-022-02807-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC), as a highly lethal malignancy with high mortality, lacks of effective treatment. Canonical therapeutic targets in PDAC demand further verification among which HER2 receptor tyrosine kinase inhibitor pyrotinib as treatment targets has not be decided. METHODS Anti-PDAC efficacy of pyrotinib was evaluated both in vitro and in vivo using both cell lines and patient-derived xenografts. By screening a large-scale library of 1453 compounds, we identified HDACs/mTOR inhibitor 1 as a promising candidate to synergize with pyrotinib. The combination therapy was evaluated in vitro and in vivo in multiple cell lines and animal models. Furthermore, RNA-seq analysis was performed to reveal the latent molecular mechanism of combination therapy. RESULTS In our study, pyrotinib monotherapy was found to be inefficient to anti-PDAC which exhibited limited anti-proliferation effect in vitro and in vivo. Through therapy combined with HDACs/mTOR inhibitor 1, pyrotinib triggered intense apoptosis in PDAC both in cell lines and animal models. Mechanistic analyses revealed that mutant P53 degradation mediated by HDAC inhibition synergized with HER2 and mTOR inhibition. CONCLUSIONS In conclusion, identification of HDACs/mTOR inhibitor as a synergistic inhibitor, provides a potent therapeutic strategy that targets HER2-positive pancreatic cancer.
Collapse
Affiliation(s)
- Tiebo Mao
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related GenesDepartment of OncologySchool of Medicine, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University, Shanghai, China
| | - Xiaofei Zhang
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related GenesDepartment of OncologySchool of Medicine, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University, Shanghai, China
| | - Haiyan Xu
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related GenesDepartment of OncologySchool of Medicine, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University, Shanghai, China
| | - Xiao Zhang
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related GenesDepartment of OncologySchool of Medicine, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University, Shanghai, China
| | - Weiyu Ge
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related GenesDepartment of OncologySchool of Medicine, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University, Shanghai, China
| | - Shumin Li
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related GenesDepartment of OncologySchool of Medicine, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University, Shanghai, China
| | - Jingyu Ma
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related GenesDepartment of OncologySchool of Medicine, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University, Shanghai, China
| | - Ming Yue
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related GenesDepartment of OncologySchool of Medicine, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University, Shanghai, China
| | - Shengbai Xue
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related GenesDepartment of OncologySchool of Medicine, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University, Shanghai, China
| | - Jiujie Cui
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related GenesDepartment of OncologySchool of Medicine, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University, Shanghai, China
| | - Liwei Wang
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related GenesDepartment of OncologySchool of Medicine, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
9
|
Turpin A, Neuzillet C, Colle E, Dusetti N, Nicolle R, Cros J, de Mestier L, Bachet JB, Hammel P. Therapeutic advances in metastatic pancreatic cancer: a focus on targeted therapies. Ther Adv Med Oncol 2022; 14:17588359221118019. [PMID: 36090800 PMCID: PMC9459481 DOI: 10.1177/17588359221118019] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/18/2022] [Indexed: 12/12/2022] Open
Abstract
Mortality from pancreatic ductal adenocarcinoma (PDAC) is increasing worldwide and effective new treatments are urgently needed. The current treatment of metastatic PDAC in fit patients is based on two chemotherapy combinations (FOLFIRINOX and gemcitabine plus nab-paclitaxel) which were validated more than 8 years ago. Although almost all treatments targeting specific molecular alterations have failed so far when administered to unselected patients, encouraging results were observed in the small subpopulations of patients with germline BRCA 1/2 mutations, and somatic gene fusions (neurotrophic tyrosine receptor kinase, Neuregulin 1, which are enriched in KRAS wild-type PDAC), KRAS G12C mutations, or microsatellite instability. While targeted tumor metabolism therapies and immunotherapy have been disappointing, they are still under investigation in combination with other drugs. Optimizing pharmacokinetics and adapting available chemotherapies based on molecular signatures are other promising avenues of research. This review evaluates the current expectations and limits of available treatments and analyses the existing trials. A permanent search for actionable vulnerabilities in PDAC tumor cells and microenvironments will probably result in a more personalized therapeutic approach, keeping in mind that supportive care must also play a major role if real clinical efficacy is to be achieved in these patients.
Collapse
Affiliation(s)
- Anthony Turpin
- Department of Medical Oncology, CNRS UMR9020,
Inserm UMR-S 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to
Therapies, University Lille, CHU Lille, Lille, France
| | - Cindy Neuzillet
- Department of Medical Oncology, Curie
Institute, Versailles Saint-Quentin University, Paris-Saclay University,
Saint-Cloud, France
| | - Elise Colle
- Department of Digestive and Medical Oncology,
Hospital Paul Brousse (AP-HP), Villejuif, University of Paris Saclay,
France
| | - Nelson Dusetti
- Cancer Research Center of Marseille, CRCM,
Inserm, CNRS, Paoli-Calmettes Institut, Aix-Marseille University, Marseille,
France
| | - Rémy Nicolle
- Centre de Recherche sur l’Inflammation, INSERM,
U1149, CNRS, ERL 8252, Université de Paris Cité, Paris, France
| | - Jérôme Cros
- Department of Pathology, University of Paris
Cité, Hospital Beaujon (AP-HP), Clichy, France
| | - Louis de Mestier
- Department of Gastroenterology and
Pancreatology, University of Paris Cité, Hospital Beaujon (AP-HP), Clichy,
France
| | - Jean-Baptiste Bachet
- Department of Gastroenterology and Digestive
Oncology, Pitié-Salpêtrière Hospital, Sorbonne University, UPMC University,
Paris, France
| | - Pascal Hammel
- Department of Digestive and Medical Oncology,
Hôpital Paul Brousse (AP-HP), 12 Avenue Paul Vaillant-Couturier, Villejuif
94800, University of Paris Saclay, France
| |
Collapse
|
10
|
O'Kane GM, Lowery MA. Moving the Needle on Precision Medicine in Pancreatic Cancer. J Clin Oncol 2022; 40:2693-2705. [PMID: 35839440 DOI: 10.1200/jco.21.02514] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 03/20/2022] [Accepted: 04/18/2022] [Indexed: 12/21/2022] Open
Abstract
The management of pancreatic ductal adenocarcinoma (PDAC) has posed a considerable challenge for decades, with incidence and mortality rates almost mirroring each other. Despite this, a deeper understanding of the complex biology inherent to PDAC has provided a roadmap for a more precise approach to treatment. PDAC deficient in homologous recombination repair and mismatch repair is a subgroup that should be identified in the clinic for a targeted approach. In addition, KRAS wild-type PDAC, occurring in approximately 10% of patients, is enriched in highly actionable alterations including fusions, underscoring the importance of integrative germline and somatic sequencing. Comprehensive sequencing efforts over the past decade have documented genomic- and transcriptomic-based classifiers, with the latter emerging as two main subtypes: the classical and basal-like, which are now being evaluated in clinical trials. Together with promising, innovative strategies to target KRAS mutations and their pleotropic effects, a new era of precision medicine in PDAC is on the horizon.
Collapse
Affiliation(s)
- Grainne M O'Kane
- Trinity St James Cancer Institute, Dublin, Ireland
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | |
Collapse
|
11
|
Ramarao-Milne P, Kondrashova O, Patch AM, Nones K, Koufariotis LT, Newell F, Addala V, Lakis V, Holmes O, Leonard C, Wood S, Xu Q, Mukhopadhyay P, Naeini MM, Steinfort D, Williamson JP, Bint M, Pahoff C, Nguyen PT, Twaddell S, Arnold D, Grainge C, Basirzadeh F, Fielding D, Dalley AJ, Chittoory H, Simpson PT, Aoude LG, Bonazzi VF, Patel K, Barbour AP, Fennell DA, Robinson BW, Creaney J, Hollway G, Pearson JV, Waddell N. Comparison of actionable events detected in cancer genomes by whole-genome sequencing, in silico whole-exome and mutation panels. ESMO Open 2022; 7:100540. [PMID: 35849877 PMCID: PMC9463385 DOI: 10.1016/j.esmoop.2022.100540] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 06/07/2022] [Accepted: 06/19/2022] [Indexed: 12/14/2022] Open
Abstract
Background Next-generation sequencing is used in cancer research to identify somatic and germline mutations, which can predict sensitivity or resistance to therapies, and may be a useful tool to reveal drug repurposing opportunities between tumour types. Multigene panels are used in clinical practice for detecting targetable mutations. However, the value of clinical whole-exome sequencing (WES) and whole-genome sequencing (WGS) for cancer care is less defined, specifically as the majority of variants found using these technologies are of uncertain significance. Patients and methods We used the Cancer Genome Interpreter and WGS in 726 tumours spanning 10 cancer types to identify drug repurposing opportunities. We compare the ability of WGS to detect actionable variants, tumour mutation burden (TMB) and microsatellite instability (MSI) by using in silico down-sampled data to mimic WES, a comprehensive sequencing panel and a hotspot mutation panel. Results We reveal drug repurposing opportunities as numerous biomarkers are shared across many solid tumour types. Comprehensive panels identify the majority of approved actionable mutations, with WGS detecting more candidate actionable mutations for biomarkers currently in clinical trials. Moreover, estimated values for TMB and MSI vary when calculated from WGS, WES and panel data, and are dependent on whether all mutations or only non-synonymous mutations were used. Our results suggest that TMB and MSI thresholds should not only be tumour-dependent, but also be sequencing platform-dependent. Conclusions There is a large opportunity to repurpose cancer drugs, and these data suggest that comprehensive sequencing is an invaluable source of information to guide clinical decisions by facilitating precision medicine and may provide a wealth of information for future studies. Furthermore, the sequencing and analysis approach used to estimate TMB may have clinical implications if a hard threshold is used to indicate which patients may respond to immunotherapy. Genome analysis revealed that treatment biomarkers are shared across solid tumours, highlighting repurposing opportunities. Comprehensive panels detect most known biomarkers; however, WGS detects more biomarkers for treatments in clinical trials. TMB is well correlated between sequencing methods, but absolute values vary and are dependent on mutation types considered.
Collapse
Affiliation(s)
- P Ramarao-Milne
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia; Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation, Brisbane, Australia
| | - O Kondrashova
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - A-M Patch
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - K Nones
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - L T Koufariotis
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - F Newell
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - V Addala
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - V Lakis
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - O Holmes
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - C Leonard
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - S Wood
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Q Xu
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - P Mukhopadhyay
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - M M Naeini
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - D Steinfort
- Department of Thoracic Medicine, Royal Melbourne Hospital, Melbourne, Australia
| | - J P Williamson
- Department of Thoracic Medicine, Liverpool Hospital Sydney, Sydney, Australia
| | - M Bint
- Department of Thoracic Medicine, Sunshine Coast University Hospital, Birtinya, Australia
| | - C Pahoff
- Department of Respiratory Medicine, Gold Coast University Hospital, Southport, Australia
| | - P T Nguyen
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia
| | - S Twaddell
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - D Arnold
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - C Grainge
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - F Basirzadeh
- Department of Thoracic Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - D Fielding
- Department of Thoracic Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - A J Dalley
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - H Chittoory
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - P T Simpson
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - L G Aoude
- The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - V F Bonazzi
- The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - K Patel
- The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - A P Barbour
- The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, Australia; Upper Gastro-intestinal Surgical Unit, Department of Surgery, Princess Alexandra Hospital, Brisbane, Australia
| | - D A Fennell
- Cancer Research UK Centre Leicester, University of Leicester & University Hospitals of Leicester NHS Trust, Leicester, UK
| | - B W Robinson
- National Centre for Asbestos Related Disease, Institute of Respiratory Health, University of Western Australia, Nedlands, Australia; Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, Australia
| | - J Creaney
- National Centre for Asbestos Related Disease, Institute of Respiratory Health, University of Western Australia, Nedlands, Australia; Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, Australia
| | - G Hollway
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - J V Pearson
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - N Waddell
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia.
| |
Collapse
|
12
|
Hirokawa YS, Iwata T, Okugawa Y, Tanaka K, Sakurai H, Watanabe M. HER2-positive adenocarcinoma arising from heterotopic pancreas tissue in the duodenum: A case report. World J Gastroenterol 2021; 27:4738-4745. [PMID: 34366633 PMCID: PMC8326258 DOI: 10.3748/wjg.v27.i28.4738] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/15/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Adenocarcinoma originating from heterotopic pancreas tissue is a rare disease. Furthermore, to our knowledge, no HER2-positive cases in the duodenum have been reported in the scientific literature nor has the efficacy of trastuzumab treatment for the disease been reported.
CASE SUMMARY A 65-year-old woman whose clinical diagnosis was unresectable advanced duodenal cancer with HER2 overexpression responded well to trastuzumab chemotherapy. The main tumor in the duodenum reduced drastically. The patient underwent pancreaticoduodenectomy and lymph node dissection. A small number of cancer cells remained in the submucosal layer of the duodenum and pancreas head. After histological and immunohistochemical examination, the patient was diagnosed with duodenal adenocarcinoma originating from heterotopic pancreas tissue.
CONCLUSION Trastuzumab treatment is effective in HER2-positive adenocarcinoma originating from heterotopic pancreas tissue in the duodenum.
Collapse
Affiliation(s)
- Yoshifumi S Hirokawa
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Mie 5148507, Japan
| | - Takashi Iwata
- Cancer Center, Aichi Medical University, Aichi 4801195, Japan
| | - Yoshinaga Okugawa
- Department of Genomic Medicine, Mie University Hospital, Mie 5148507, Japan
| | - Koji Tanaka
- Department of Surgery, IGA City General Hospital, Mie 5180823, Japan
| | - Hiroyuki Sakurai
- Hepato-biliary-pancreatic and Transplant Surgery, Mie University Hospital, Mie 5148507, Japan
| | - Masatoshi Watanabe
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Mie 5148507, Japan
| |
Collapse
|
13
|
Karelia DN, Kim S, K. Pandey M, Plano D, Amin S, Lu J, Sharma AK. Novel Seleno-Aspirinyl Compound AS-10 Induces Apoptosis, G1 Arrest of Pancreatic Ductal Adenocarcinoma Cells, Inhibits Their NF-κB Signaling, and Synergizes with Gemcitabine Cytotoxicity. Int J Mol Sci 2021; 22:4966. [PMID: 34067020 PMCID: PMC8124556 DOI: 10.3390/ijms22094966] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023] Open
Abstract
Current available therapies for pancreatic ductal adenocarcinoma (PDAC) provide minimal overall survival benefits and cause severe adverse effects. We have identified a novel molecule AS-10, a selenazolidine-bis-aspirinyl derivative, that was two to three orders of magnitude more potent than aspirin and at least one to two orders of magnitude more potent than gemcitabine in inhibiting PDAC cancer cell growth/viability against three PDAC cell lines while sparing mouse embryonic fibroblasts in the same exposure range. In Panc-1 cells, AS-10 induced apoptosis without necrosis, principally through caspase-3/7 cascade and reactive oxygen species, in addition to an induction of G1 cell cycle block. Transcriptomic profiling with RNA-seq indicated the top responses to AS-10 exposure as CDKN1A (P21Cip1), CCND1, and nuclear transcription factor-kappa B (NF-κB) complex and the top functions as cell cycle, cell death, and survival without inducing the DNA damage gene signature. AS-10 pretreatment (6 h) decreased cytokine tumor necrosis factor-alpha (TNF-α)-stimulated NF-κB nuclear translocation, DNA binding activity, and degradation of cytosolic inhibitor of κB (IκB) protein. As NF-κB activation in PDAC cells confers resistance to gemcitabine, the AS-10 combination with gemcitabine increased the in vitro cytotoxicity more than the additivity of both compounds. Overall, our results suggest AS-10 may be a promising drug lead for PDAC, both as a single agent and in combination therapy.
Collapse
Affiliation(s)
- Deepkamal N. Karelia
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA; (D.N.K.); (S.K.); (M.K.P.); (D.P.); (S.A.)
| | - Sangyub Kim
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA; (D.N.K.); (S.K.); (M.K.P.); (D.P.); (S.A.)
| | - Manoj K. Pandey
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA; (D.N.K.); (S.K.); (M.K.P.); (D.P.); (S.A.)
| | - Daniel Plano
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA; (D.N.K.); (S.K.); (M.K.P.); (D.P.); (S.A.)
| | - Shantu Amin
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA; (D.N.K.); (S.K.); (M.K.P.); (D.P.); (S.A.)
- Penn State Cancer Institute, 500 University Drive, Hershey, PA 17033, USA
| | - Junxuan Lu
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA; (D.N.K.); (S.K.); (M.K.P.); (D.P.); (S.A.)
- Penn State Cancer Institute, 500 University Drive, Hershey, PA 17033, USA
| | - Arun K. Sharma
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA; (D.N.K.); (S.K.); (M.K.P.); (D.P.); (S.A.)
- Penn State Cancer Institute, 500 University Drive, Hershey, PA 17033, USA
| |
Collapse
|
14
|
Mechanisms of drug resistance of pancreatic ductal adenocarcinoma at different levels. Biosci Rep 2021; 40:225827. [PMID: 32677676 PMCID: PMC7396420 DOI: 10.1042/bsr20200401] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 07/05/2020] [Accepted: 07/16/2020] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related death worldwide, and the mortality of patients with PDAC has not significantly decreased over the last few decades. Novel strategies exhibiting promising effects in preclinical or phase I/II clinical trials are often situated in an embarrassing condition owing to the disappointing results in phase III trials. The efficacy of the current therapeutic regimens is consistently compromised by the mechanisms of drug resistance at different levels, distinctly more intractable than several other solid tumours. In this review, the main mechanisms of drug resistance clinicians and investigators are dealing with during the exploitation and exploration of the anti-tumour effects of drugs in PDAC treatment are summarized. Corresponding measures to overcome these limitations are also discussed.
Collapse
|
15
|
Liu X, Li Z, Wang Y. Advances in Targeted Therapy and Immunotherapy for Pancreatic Cancer. Adv Biol (Weinh) 2021; 5:e1900236. [PMID: 33729700 DOI: 10.1002/adbi.201900236] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 08/19/2020] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer is a highly aggressive malignancy with an overall 5-year survival rate of <6% due to therapeutic resistance and late-stage diagnosis. These statistics have not changed despite 50 years of research and therapeutic development. Pancreatic cancer is predicted to become the second leading cause of cancer mortality by the year 2030. Currently, the treatment options for pancreatic cancer are limited. This disease is usually diagnosed at a late stage, which prevents curative surgical resection. Chemotherapy is the most frequently used approach for pancreatic cancer treatment and has limited effects. In many other cancer types, targeted therapy and immunotherapy have made great progress and have been shown to be very promising prospects; these treatments also provide hope for pancreatic cancer. The need for research on targeted therapy and immunotherapy is pressing due to the poor prognosis of pancreatic cancer, and in recent years, there have been some breakthroughs for targeted therapy and immunotherapy in pancreatic cancer. This review summarizes the current preclinical and clinical studies of targeted therapy and immunotherapy for pancreatic cancer and ends by describing the challenges and outlook.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, SINH - Changzheng Hospital Joint Center for Translational Medicine, Institutes for Translational Medicine (CAS-SMMU), Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhang Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, SINH - Changzheng Hospital Joint Center for Translational Medicine, Institutes for Translational Medicine (CAS-SMMU), Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yuexiang Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, SINH - Changzheng Hospital Joint Center for Translational Medicine, Institutes for Translational Medicine (CAS-SMMU), Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
16
|
Luo Y, Hu J, Liu Y, Li L, Li Y, Sun B, Kong R. Invadopodia: A potential target for pancreatic cancer therapy. Crit Rev Oncol Hematol 2021; 159:103236. [PMID: 33482351 DOI: 10.1016/j.critrevonc.2021.103236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 01/05/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023] Open
Abstract
Dissemination of cancer cells is an intricate multistep process that represents the most deadly aspect of cancer. Cancer cells form F-actin-rich protrusions known as invadopodia to invade surrounding tissues, blood vessels and lymphatics. A number of studies have demonstrated the significant roles of invadopodia in cancer. Therefore, the specific cells and molecules involved in invadopodia activity can provide as therapeutic targets. In this review, we included a thorough overview of studies in invadopodia and discussed their functions in cancer metastasis. We then presented the specific cells and molecules involved in invadopodia activity in pancreatic cancer and analyzed their suitability to be effective therapeutic targets. Currently, drugs targeting invadopodia and relevant clinical trials are negligible. Here, we highlighted the significance of potential drugs and discussed future obstacles in implementing clinical trials. This review presents a new perspective on invadopodia-induced pancreatic cancer metastasis and may prosper the development of targeted therapeutics against pancreatic cancer.
Collapse
Affiliation(s)
- Yan Luo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jisheng Hu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Kong
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
17
|
Froeling FEM, Casolino R, Pea A, Biankin AV, Chang DK. Molecular Subtyping and Precision Medicine for Pancreatic Cancer. J Clin Med 2021; 10:E149. [PMID: 33406790 PMCID: PMC7794969 DOI: 10.3390/jcm10010149] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/13/2020] [Accepted: 12/25/2020] [Indexed: 12/12/2022] Open
Abstract
Substantial progress in recent years has dramatically increased our knowledge of the molecular basis of cancer, revealing new potential therapeutic targets and paving the way for effective personalised medicine for the treatment of many tumour types. However, pancreatic cancer has been lagging behind in this success and continues to be one of the most lethal solid malignancies. Its molecular heterogeneity and the unselected design of the majority of clinical trials to date can in part explain the reason for our failure to make a significant change in the survival outcomes for patients with pancreatic cancer. A changing paradigm in drug development is required to validate the new molecular taxonomy and to rapidly translate preclinical discovery into clinical trials. Here, we review the molecular subtyping of pancreatic cancer, the challenges in identifying effective treatment regimens according to defined low-prevalence molecular subgroups and we illustrate a new model of translational therapeutic development that was established in the U.K. (Precision-Panc) as a potentially effective solution to improve outcomes for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Fieke E. M. Froeling
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; (F.E.M.F.); (R.C.); (A.P.); (A.V.B.)
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Raffaella Casolino
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; (F.E.M.F.); (R.C.); (A.P.); (A.V.B.)
- Department of Medicine, University and Hospital Trust of Verona of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Antonio Pea
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; (F.E.M.F.); (R.C.); (A.P.); (A.V.B.)
- Department of Surgery, University and Hospital Trust of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Andrew V. Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; (F.E.M.F.); (R.C.); (A.P.); (A.V.B.)
| | - David K. Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; (F.E.M.F.); (R.C.); (A.P.); (A.V.B.)
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow G31 2ER, UK
| | | |
Collapse
|
18
|
Lengyel CG, Habeeb B, Khan SZ, El Bairi K, Altuna SC, Hussain S, Mazher SA, Trapani D, Petrillo A. Role of Her-2 in Gastrointestinal Tumours beyond Gastric Cancer: A Tool for Precision Medicine. GASTROINTESTINAL DISORDERS 2020; 3:1-22. [DOI: 10.3390/gidisord3010001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal (GI) tumors account for a quarter of all the cancer burden and a third of the global cancer-related mortality. Among them, some cancers retain a dismal prognosis; therefore, newer and innovative therapies are urgently needed in priority disease areas of high-unmet medical need. In this context, HER2 could be a relevant prognostic and predictive biomarker acting as a target for specific drugs. However, if the role of HER2 has been object of investigation for several years in gastric cancer, it is not well established in other GI malignancies. The aim of this narrative review was to portray the current landscape of the potential role of HER2 as a predictive biomarker for GI tumors beyond gastric cancer. In colon cancer, the benefit from anti-HER2 therapies is less clear than in gastric neoplasms for the lack of controlled studies. Pancreatic, biliary tract adenocarcinomas and hepatocarcinoma may derive a less clear clinical benefit by using anti-HER2 agents in HER2 positive tumors. Overall, the results are promising and seem to suggest that the integration of multiple modalities of therapies can optimize the cancer care. However, further prospective trials are needed to validate the use of personalized targeted therapies in this field.
Collapse
Affiliation(s)
- Csongor G. Lengyel
- Head and Neck Surgery, National Institute of Oncology, 1122 Budapest, Hungary
| | - Baker Habeeb
- Medical Oncology Department, Shaqlawa Teaching Hospital, Erbil 44001, Iraq
| | - Shah Z. Khan
- Department of Clinical Oncology, BINOR Cancer Hospital, Bannu 28100, Pakistan
| | | | | | - Sadaqat Hussain
- Northwest Cancer Center, Western Health and Social Care Trust, Altnagelvin Hospital, Londonderry BT47 6SB, UK
| | - Syed Ayub Mazher
- UT Southwestern Clements University Hospital, Dallas, TX 75390, USA
| | - Dario Trapani
- European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Angelica Petrillo
- Medical Oncology Unit, Ospedale del Mare, 80147 Naples, Italy
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Study of Campania “L.Vanvitelli”, 81100 Caserta, Italy
| |
Collapse
|
19
|
Assenat E, Mineur L, Mollevi C, Lopez-Crapez E, Lombard-Bohas C, Samalin E, Portales F, Walter T, de Forges H, Dupuy M, Boissière-Michot F, Ho-Pun-Cheung A, Ychou M, Mazard T. Phase II study evaluating the association of gemcitabine, trastuzumab and erlotinib as first-line treatment in patients with metastatic pancreatic adenocarcinoma (GATE 1). Int J Cancer 2020; 148:682-691. [PMID: 33405269 DOI: 10.1002/ijc.33225] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022]
Abstract
In a previous phase II study (THERAPY), cetuximab and trastuzumab combination, as second-line after progression with gemcitabine, showed disease stabilization in 27% of 33 patients with pancreatic carcinoma. In the present phase II multicenter study, we assessed the efficacy and tolerance of gemcitabine, trastuzumab plus erlotinib as first-line treatment of metastatic pancreatic cancer. The primary endpoint was disease control rate (DCR, RECIST v.1); secondary endpoints were progression-free (PFS), overall (OS) survival and toxicity (NCI-CTCAE v3.0). Ancillary study addressed the predictive value of both EGFR/HER2 expression and KRAS mutational status. Sixty-three patients from four centers were included (62 evaluable for toxicity, 59 for efficacy), median age was 62 years (35-77), 59.7% men. The median treatment duration was 16.1 weeks (2.1-61). Eleven patients (19%) reported a partial tumor response, and 33 (56%) disease stabilization. DCR was 74.6% (95%CI: 61.8-85.0; 44/59 patients). After a median follow-up of 23.3 months (0.6-23.6), median PFS was 3.5 months (95%CI: 2.4-3.8) and median OS 7.9 months (95%CI: 5.1-10.2). PFS was significantly longer in patients with grade ≥ 2 cutaneous toxicities vs patients with grade 0-1 toxicities (HR = 0.55, 95%CI: 0.33-0.92, P = .020). Expression of EGFR and HER2 was correlated with PFS and OS in multivariate analysis; HER2 expression was correlated with the tumor response. Main severe toxicities were neutropenia (32%), cutaneous rash (37%) and thrombosis/embolisms (35.5%). This triplet combination is effective in terms of disease control, PFS and OS, and acceptable for safety. A larger study to investigate this combination compared to the standard regimen should be discussed.
Collapse
Affiliation(s)
- Eric Assenat
- Medical Oncology Department, Institut du Cancer de Montpellier (ICM), Univ. Montpellier, Montpellier, France.,Centre Hospitalier Régional Universitaire (CHU) de Montpellier, Univ. Montpellier, Montpellier, France
| | - Laurent Mineur
- Unité de Cancérologie Digestive Oncologie Radiothérapie, Institut Sainte Catherine, Avignon, France
| | - Caroline Mollevi
- Biometrics Unit, Institut du Cancer de Montpellier (ICM), University of Montpellier, Montpellier, France
| | - Evelyne Lopez-Crapez
- Translational Research Unit, Institut Régional du Cancer de Montpellier (ICM), University of Montpellier, Montpellier, France
| | | | - Emmanuelle Samalin
- Medical Oncology Department, Institut du Cancer de Montpellier (ICM), Univ. Montpellier, Montpellier, France
| | - Fabienne Portales
- Medical Oncology Department, Institut du Cancer de Montpellier (ICM), Univ. Montpellier, Montpellier, France
| | | | - Hélène de Forges
- Clinical Research and Innovation Department, Institut du Cancer de Montpellier (ICM), University of Montpellier, Montpellier, France
| | - Marie Dupuy
- Centre Hospitalier Régional Universitaire (CHU) de Montpellier, Univ. Montpellier, Montpellier, France
| | - Florence Boissière-Michot
- Translational Research Unit, Institut Régional du Cancer de Montpellier (ICM), University of Montpellier, Montpellier, France
| | - Alexandre Ho-Pun-Cheung
- Translational Research Unit, Institut Régional du Cancer de Montpellier (ICM), University of Montpellier, Montpellier, France
| | - Marc Ychou
- Medical Oncology Department, Institut du Cancer de Montpellier (ICM), Univ. Montpellier, Montpellier, France
| | - Thibaut Mazard
- Medical Oncology Department, Institut du Cancer de Montpellier (ICM), Univ. Montpellier, Montpellier, France
| |
Collapse
|
20
|
Subasinghe D, Acott N, Kumarasinghe MP. A survival guide to HER2 testing in gastric/gastroesophageal junction carcinoma. Gastrointest Endosc 2019; 90:44-54. [PMID: 30928424 DOI: 10.1016/j.gie.2019.03.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 03/14/2019] [Indexed: 02/06/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) status determines gastric/gastroesophageal junction (GEJ) adenocarcinomas that benefit from targeted therapy; hence, HER2 testing has become a routine practice. Accurate HER2 testing is fundamental to select eligible patients who will benefit from HER2-targeted treatment. The reported HER2-positive rate in gastric/GEJ cancers ranges from 4.4% to 53.4%, and HER2-positive tumors are considered to have more-aggressive biologic behavior and tumor recurrence. Main modalities of HER2 testing in clinical practice include immunohistochemistry (IHC) for protein expression and in situ hybridization (ISH) for gene amplification. Many technical pitfalls affect the accuracy of HER2 result. Additionally, several issues in HER2 testing are related to the tumor biology, sample selection, interpretation of IHC and ISH results, and confirming HER2 status. Therefore, gastric/GEJ adenocarcinoma-specific HER2 testing protocols have been developed and standardized to minimize the impact of these preanalytical and analytical factors and to enhance reproducibility of HER2 testing results. This review provides up-to-date practical guidance to clinicians on accurate HER2 testing and interpretation of results in gastric/GEJ adenocarcinoma.
Collapse
Affiliation(s)
- Duminda Subasinghe
- Department of Surgery, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka; Digestive Disease Unit, Aintree University Hospital, NHS Foundation Trust, Liverpool, UK; Pathwest Laboratory Medicine, Perth and University of Western Australia, Perth, Australia
| | - Nathan Acott
- Pathwest Laboratory Medicine, Perth and University of Western Australia, Perth, Australia
| | | |
Collapse
|
21
|
Bourillon L, Bourgier C, Gaborit N, Garambois V, Llès E, Zampieri A, Ogier C, Jarlier M, Radosevic-Robin N, Orsetti B, Delpech H, Theillet C, Colombo PE, Azria D, Pèlegrin A, Larbouret C, Chardès T. An auristatin-based antibody-drug conjugate targeting HER3 enhances the radiation response in pancreatic cancer. Int J Cancer 2019; 145:1838-1851. [PMID: 30882895 DOI: 10.1002/ijc.32273] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/20/2019] [Accepted: 03/01/2019] [Indexed: 12/17/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer characterized by poor response to chemotherapy and radiotherapy due to the lack of efficient therapeutic tools and early diagnostic markers. We previously generated the nonligand competing anti-HER3 antibody 9F7-F11 that binds to pancreatic tumor cells and induces tumor regression in vivo in experimental models. Here, we asked whether coupling 9F7-F11 with a radiosensitizer, such as monomethylauristatin E (MMAE), by using the antibody-drug conjugate (ADC) technology could improve radiation therapy efficacy in PDAC. We found that the MMAE-based HER3 antibody-drug conjugate (HER3-ADC) was efficiently internalized in tumor cells, increased the fraction of cells arrested in G2/M, which is the most radiosensitive phase of the cell cycle, and promoted programmed cell death of irradiated HER3-positive pancreatic cancer cells (BxPC3 and HPAC cell lines). HER3-ADC decreased the clonogenic survival of irradiated cells by increasing DNA double-strand break formation (based on γH2AX level), and by modulating DNA damage repair. Tumor radiosensitization with HER3-ADC favored the inhibition of the AKT-induced survival pathway, together with more efficient caspase 3/PARP-mediated apoptosis. Incubation with HER3-ADC before irradiation synergistically reduced the phosphorylation of STAT3, which is involved in chemoradiation resistance. In vivo, the combination of HER3-ADC with radiation therapy increased the overall survival of mice harboring BxPC3, HPAC cell xenografts or patient-derived xenografts, and reduced proliferation (KI67-positive cells). Combining auristatin radiosensitizer delivery via an HER3-ADC with radiotherapy is a new promising therapeutic strategy in PDAC.
Collapse
Affiliation(s)
- Laura Bourillon
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Céline Bourgier
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France.,Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Nadège Gaborit
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Véronique Garambois
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Eva Llès
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Alexandre Zampieri
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Charline Ogier
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Marta Jarlier
- Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Nina Radosevic-Robin
- Department of Biopathology, Jean Perrin Comprehensive Cancer Center and INSERM/UCA UMR 1240, 63011, Clermont-Ferrand, France
| | - Béatrice Orsetti
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Hélène Delpech
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Charles Theillet
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Pierre-Emmanuel Colombo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France.,Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - David Azria
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France.,Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - André Pèlegrin
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Christel Larbouret
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France
| | - Thierry Chardès
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298, Montpellier, France.,Centre National de la Recherche Scientifique (CNRS), Paris, France
| |
Collapse
|
22
|
Conway JRW, Herrmann D, Evans TRJ, Morton JP, Timpson P. Combating pancreatic cancer with PI3K pathway inhibitors in the era of personalised medicine. Gut 2019; 68:742-758. [PMID: 30396902 PMCID: PMC6580874 DOI: 10.1136/gutjnl-2018-316822] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the most deadly solid tumours. This is due to a generally late-stage diagnosis of a primarily treatment-refractory disease. Several large-scale sequencing and mass spectrometry approaches have identified key drivers of this disease and in doing so highlighted the vast heterogeneity of lower frequency mutations that make clinical trials of targeted agents in unselected patients increasingly futile. There is a clear need for improved biomarkers to guide effective targeted therapies, with biomarker-driven clinical trials for personalised medicine becoming increasingly common in several cancers. Interestingly, many of the aberrant signalling pathways in PDAC rely on downstream signal transduction through the mitogen-activated protein kinase and phosphoinositide 3-kinase (PI3K) pathways, which has led to the development of several approaches to target these key regulators, primarily as combination therapies. The following review discusses the trend of PDAC therapy towards molecular subtyping for biomarker-driven personalised therapies, highlighting the key pathways under investigation and their relationship to the PI3K pathway.
Collapse
Affiliation(s)
- James RW Conway
- Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Cancer Division, Sydney, New South Wales, Australia
| | - David Herrmann
- Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Cancer Division, Sydney, New South Wales, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - TR Jeffry Evans
- Cancer Department, Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Jennifer P Morton
- Cancer Department, Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Paul Timpson
- Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Cancer Division, Sydney, New South Wales, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
23
|
Barati Bagherabad M, Afzaljavan F, ShahidSales S, Hassanian SM, Avan A. Targeted therapies in pancreatic cancer: Promises and failures. J Cell Biochem 2018; 120:2726-2741. [PMID: 28703890 DOI: 10.1002/jcb.26284] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/11/2018] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an incidence rate nearly equal to its mortality rate. The poor prognosis of the disease can be explained by the absence of effective biomarkers for screening and early detection, together with the aggressive behavior and resistance to the currently available chemotherapy. The therapeutic failure can also be attributed to the inter-/intratumor genetic heterogeneity and the abundance of tumor stroma that occupies the majority of the tumor mass. Gemcitabine is used in the treatment of PDAC; however, the response rate is less than 12%. A recent phase III trial revealed that the combination of oxaliplatin, irinotecan, fluorouracil, and leucovorin could be an option for the treatment of metastatic PDAC patients with good performance status, although these approaches can result in high toxicity level. Further investigations are required to develop innovative anticancer agents that either improve gemcitabine activity, within novel combinatorial approaches or acts with a better efficacy than gemcitabine. The aim of the current review is to give an overview of preclinical and clinical studies targeting key dysregulated signaling pathways in PDAC.
Collapse
Affiliation(s)
- Matineh Barati Bagherabad
- Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Afzaljavan
- Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodabeh ShahidSales
- Cancer Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran.,Molecular Medicine group, Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Heining C, Horak P, Uhrig S, Codo PL, Klink B, Hutter B, Fröhlich M, Bonekamp D, Richter D, Steiger K, Penzel R, Endris V, Ehrenberg KR, Frank S, Kleinheinz K, Toprak UH, Schlesner M, Mandal R, Schulz L, Lambertz H, Fetscher S, Bitzer M, Malek NP, Horger M, Giese NA, Strobel O, Hackert T, Springfeld C, Feuerbach L, Bergmann F, Schröck E, von Kalle C, Weichert W, Scholl C, Ball CR, Stenzinger A, Brors B, Fröhling S, Glimm H. NRG1 Fusions in KRAS Wild-Type Pancreatic Cancer. Cancer Discov 2018; 8:1087-1095. [PMID: 29802158 DOI: 10.1158/2159-8290.cd-18-0036] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/24/2018] [Accepted: 05/24/2018] [Indexed: 11/16/2022]
Abstract
We used whole-genome and transcriptome sequencing to identify clinically actionable genomic alterations in young adults with pancreatic ductal adenocarcinoma (PDAC). Molecular characterization of 17 patients with PDAC enrolled in a precision oncology program revealed gene fusions amenable to pharmacologic inhibition by small-molecule tyrosine kinase inhibitors in all patients with KRAS wild-type (KRASWT) tumors (4 of 17). These alterations included recurrent NRG1 rearrangements predicted to drive PDAC development through aberrant ERBB receptor-mediated signaling, and pharmacologic ERBB inhibition resulted in clinical improvement and remission of liver metastases in 2 patients with NRG1-rearranged tumors that had proved resistant to standard treatment. Our findings demonstrate that systematic screening of KRASWT tumors for oncogenic fusion genes will substantially improve the therapeutic prospects for a sizeable fraction of patients with PDAC.Significance: Advanced PDAC is a malignancy with few treatment options that lacks molecular mechanism-based therapies. Our study uncovers recurrent gene rearrangements such as NRG1 fusions as disease-driving events in KRASwt tumors, thereby providing novel insights into oncogenic signaling and new therapeutic options in this entity. Cancer Discov; 8(9); 1087-95. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 1047.
Collapse
Affiliation(s)
- Christoph Heining
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany.,University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Dresden, Germany
| | - Peter Horak
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg and DKFZ, Heidelberg, Germany.,Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany.,DKTK, Heidelberg, Germany
| | - Sebastian Uhrig
- DKTK, Heidelberg, Germany.,Division of Applied Bioinformatics, DKFZ and NCT Heidelberg, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Paula L Codo
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Barbara Klink
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Dresden, Germany.,Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Barbara Hutter
- DKTK, Heidelberg, Germany.,Division of Applied Bioinformatics, DKFZ and NCT Heidelberg, Heidelberg, Germany
| | - Martina Fröhlich
- DKTK, Heidelberg, Germany.,Division of Applied Bioinformatics, DKFZ and NCT Heidelberg, Heidelberg, Germany
| | | | - Daniela Richter
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Dresden, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University Munich, Munich, Germany.,DKTK, Munich, Germany
| | - Roland Penzel
- DKTK, Heidelberg, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Volker Endris
- DKTK, Heidelberg, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Karl Roland Ehrenberg
- Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.,Department of Medical Oncology, NCT, Heidelberg, Germany
| | - Stephanie Frank
- Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Kortine Kleinheinz
- Division of Theoretical Bioinformatics, DKFZ, Heidelberg, Germany.,Department for Bioinformatics and Functional Genomics, Institute for Pharmacy and Molecular Biotechnology and BioQuant, Heidelberg University, Heidelberg, Germany
| | - Umut H Toprak
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.,Division of Theoretical Bioinformatics, DKFZ, Heidelberg, Germany.,Bioinformatics and Omics Data Analytics, DKFZ, Heidelberg, Germany
| | | | - Ranadip Mandal
- Division of Applied Functional Genomics, DKFZ, Heidelberg, Germany
| | - Lothar Schulz
- Department of Oncology, Klinikum Garmisch-Partenkirchen, Garmisch-Partenkirchen, Germany
| | - Helmut Lambertz
- Department of Oncology, Klinikum Garmisch-Partenkirchen, Garmisch-Partenkirchen, Germany
| | | | - Michael Bitzer
- Department of Gastroenterology, Hepatology and Infectious Diseases, Tübingen University Hospital, Tübingen, Germany.,DKTK, Tübingen, Germany
| | - Nisar P Malek
- Department of Gastroenterology, Hepatology and Infectious Diseases, Tübingen University Hospital, Tübingen, Germany.,DKTK, Tübingen, Germany
| | - Marius Horger
- DKTK, Tübingen, Germany.,Department of Radiology, Tübingen University Hospital, Tübingen, Germany
| | - Nathalia A Giese
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Oliver Strobel
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thilo Hackert
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Lars Feuerbach
- DKTK, Heidelberg, Germany.,Division of Applied Bioinformatics, DKFZ and NCT Heidelberg, Heidelberg, Germany
| | - Frank Bergmann
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Evelin Schröck
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Dresden, Germany.,Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Christof von Kalle
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg and DKFZ, Heidelberg, Germany.,Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany.,DKTK, Heidelberg, Germany.,DKFZ-Heidelberg Center for Personalized Oncology (HIPO), Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University Munich, Munich, Germany.,DKTK, Munich, Germany
| | - Claudia Scholl
- DKTK, Heidelberg, Germany.,Division of Applied Functional Genomics, DKFZ, Heidelberg, Germany
| | - Claudia R Ball
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Albrecht Stenzinger
- DKTK, Heidelberg, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Benedikt Brors
- DKTK, Heidelberg, Germany.,Division of Applied Bioinformatics, DKFZ and NCT Heidelberg, Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg and DKFZ, Heidelberg, Germany. .,Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany.,DKTK, Heidelberg, Germany.,Department of Medical Oncology, NCT, Heidelberg, Germany
| | - Hanno Glimm
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany. .,University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Dresden, Germany
| |
Collapse
|
25
|
Tesfaye AA, Kamgar M, Azmi A, Philip PA. The evolution into personalized therapies in pancreatic ductal adenocarcinoma: challenges and opportunities. Expert Rev Anticancer Ther 2018; 18:131-148. [PMID: 29254387 PMCID: PMC6121777 DOI: 10.1080/14737140.2018.1417844] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 12/12/2017] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is projected to be the second leading cause of cancer related mortality in the United States in 2030, with a 5-year overall survival of less than 10% despite decades of extensive research. Pancreatic cancer is marked by the accumulation of complex molecular changes, complex tumor-stroma interaction, and an immunosuppressive tumor microenvironment. PDAC has proven to be resistant to many cytotoxic, targeted and immunologic treatment approaches. Areas covered: In this paper, we review the major areas of research in PDAC, with highlights on the challenges and areas of opportunity for personalized treatment approaches. Expert commentary: The focus of research in pancreatic cancer has moved away from developing conventional cytotoxic combinations. The marked advances in understanding the molecular biology of this disease especially in the areas of the microenvironment, metabolism, and DNA repair have opened new opportunities for developing novel treatment strategies. Improved understanding of molecular abnormalities allows the development of personalized treatment approaches.
Collapse
Affiliation(s)
- Anteneh A Tesfaye
- Department of Oncology, Wayne State University, School of Medicine, Detroit, MI
- Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - Mandana Kamgar
- Department of Oncology, Wayne State University, School of Medicine, Detroit, MI
- Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - Asfar Azmi
- Department of Oncology, Wayne State University, School of Medicine, Detroit, MI
- Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - Philip A Philip
- Department of Oncology, Wayne State University, School of Medicine, Detroit, MI
- Barbara Ann Karmanos Cancer Institute, Detroit, MI
- Department of Pharmacology, Wayne State University, School of Medicine, Detroit, MI
| |
Collapse
|
26
|
Chiaravalli M, Reni M, O'Reilly EM. Pancreatic ductal adenocarcinoma: State-of-the-art 2017 and new therapeutic strategies. Cancer Treat Rev 2017; 60:32-43. [DOI: 10.1016/j.ctrv.2017.08.007] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 08/13/2017] [Accepted: 08/14/2017] [Indexed: 12/18/2022]
|
27
|
Malgerud L, Lindberg J, Wirta V, Gustafsson‐Liljefors M, Karimi M, Moro CF, Stecker K, Picker A, Huelsewig C, Stein M, Bohnert R, Del Chiaro M, Haas SL, Heuchel RL, Permert J, Maeurer MJ, Brock S, Verbeke CS, Engstrand L, Jackson DB, Grönberg H, Löhr J. Bioinformatory-assisted analysis of next-generation sequencing data for precision medicine in pancreatic cancer. Mol Oncol 2017; 11:1413-1429. [PMID: 28675654 PMCID: PMC5623817 DOI: 10.1002/1878-0261.12108] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/30/2017] [Accepted: 06/10/2017] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a tumor with an extremely poor prognosis, predominantly as a result of chemotherapy resistance and numerous somatic mutations. Consequently, PDAC is a prime candidate for the use of sequencing to identify causative mutations, facilitating subsequent administration of targeted therapy. In a feasibility study, we retrospectively assessed the therapeutic recommendations of a novel, evidence-based software that analyzes next-generation sequencing (NGS) data using a large panel of pharmacogenomic biomarkers for efficacy and toxicity. Tissue from 14 patients with PDAC was sequenced using NGS with a 620 gene panel. FASTQ files were fed into treatmentmap. The results were compared with chemotherapy in the patients, including all side effects. No changes in therapy were made. Known driver mutations for PDAC were confirmed (e.g. KRAS, TP53). Software analysis revealed positive biomarkers for predicted effective and ineffective treatments in all patients. At least one biomarker associated with increased toxicity could be detected in all patients. Patients had been receiving one of the currently approved chemotherapy agents. In two patients, toxicity could have been correctly predicted by the software analysis. The results suggest that NGS, in combination with an evidence-based software, could be conducted within a 2-week period, thus being feasible for clinical routine. Therapy recommendations were principally off-label use. Based on the predominant KRAS mutations, other drugs were predicted to be ineffective. The pharmacogenomic biomarkers indicative of increased toxicity could be retrospectively linked to reported negative side effects in the respective patients. Finally, the occurrence of somatic and germline mutations in cancer syndrome-associated genes is noteworthy, despite a high frequency of these particular variants in the background population. These results suggest software-analysis of NGS data provides evidence-based information on effective, ineffective and toxic drugs, potentially forming the basis for precision cancer medicine in PDAC.
Collapse
Affiliation(s)
- Linnéa Malgerud
- Center for Digestive DiseasesKarolinska University HospitalStockholmSweden
- Department of Clinical SciencesIntervention and Technology (CLINTEC)Karolinska InstitutetStockholmSweden
| | - Johan Lindberg
- Department of Medical Epidemiology & Biostatistics (MEB)Karolinska InstitutetStockholmSweden
| | - Valtteri Wirta
- Science for Life LaboratoryDepartment of Microbiology, Tumor and Cell Biology (MTC)Karolinska InstitutetStockholmSweden
| | | | - Masoud Karimi
- Department of Oncology at RadiumhemmetKarolinska University HospitalStockholmSweden
| | | | | | | | | | | | | | - Marco Del Chiaro
- Center for Digestive DiseasesKarolinska University HospitalStockholmSweden
- Department of Clinical SciencesIntervention and Technology (CLINTEC)Karolinska InstitutetStockholmSweden
| | - Stephan L. Haas
- Center for Digestive DiseasesKarolinska University HospitalStockholmSweden
| | - Rainer L. Heuchel
- Department of Clinical SciencesIntervention and Technology (CLINTEC)Karolinska InstitutetStockholmSweden
| | - Johan Permert
- Innovation OfficeKarolinska University HospitalStockholmSweden
| | - Markus J. Maeurer
- Department of Laboratory Medicine (LABMED)Karolinska InstitutetStockholmSweden
| | | | | | - Lars Engstrand
- Science for Life LaboratoryDepartment of Microbiology, Tumor and Cell Biology (MTC)Karolinska InstitutetStockholmSweden
| | | | - Henrik Grönberg
- Department of Medical Epidemiology & Biostatistics (MEB)Karolinska InstitutetStockholmSweden
| | - Johannes‐Matthias Löhr
- Center for Digestive DiseasesKarolinska University HospitalStockholmSweden
- Department of Clinical SciencesIntervention and Technology (CLINTEC)Karolinska InstitutetStockholmSweden
| |
Collapse
|
28
|
Ruess DA, Görgülü K, Wörmann SM, Algül H. Pharmacotherapeutic Management of Pancreatic Ductal Adenocarcinoma: Current and Emerging Concepts. Drugs Aging 2017; 34:331-357. [PMID: 28349415 DOI: 10.1007/s40266-017-0453-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pancreatic ductal adenocarcinoma is a devastating malignancy, which is the result of late diagnosis, aggressive disease, and a lack of effective treatment options. Thus, pancreatic ductal adenocarcinoma is projected to become the second leading cause of cancer-related death by 2030. This review summarizes recent developments of oncological therapy in the palliative setting of metastatic pancreatic ductal adenocarcinoma. It further compiles novel targets and therapeutic approaches as well as promising treatment combinations, which are presently in preclinical evaluation, covering several aspects of the hallmarks of cancer. Finally, challenges to the implementation of an individualized therapy approach in the context of precision medicine are discussed.
Collapse
Affiliation(s)
- Dietrich A Ruess
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Kivanc Görgülü
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Sonja M Wörmann
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Hana Algül
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.
| |
Collapse
|
29
|
Matera R, Saif MW. New therapeutic directions for advanced pancreatic cancer: cell cycle inhibitors, stromal modifiers and conjugated therapies. Expert Opin Emerg Drugs 2017; 22:223-233. [PMID: 28783977 DOI: 10.1080/14728214.2017.1362388] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Pancreatic adenocarcinoma is a devastating malignancy with an extremely poor prognosis. These tumors progress rapidly and somewhat silently with few specific symptoms and are relatively resistant to chemotherapeutic agents. Many agents, including cell cycle inhibitors, are under development for the treatment of this cancer for which there are disappointingly few treatment options. Areas covered: Here we outline the existing approved treatments for advanced pancreatic disease and discuss a range of novel therapies currently under development including cell cycle inhibitors, stromal modifiers and conjugated therapies. We also describe the current state of the pancreatic cancer therapeutics market both past and future. Expert opinion: Despite the recent explosion of novel therapies with an array of unique targets, the core treatment of pancreatic cancer still with traditional cytotoxic agents with a few exceptions. However, as these novel treatments move through the pipeline, we are hopeful that there will soon be a number of effective options for patients with advanced pancreatic cancer.
Collapse
Affiliation(s)
- Robert Matera
- a Department of Hematology and Oncology , Tufts University School of Medicine , Boston , MA , USA
| | - Muhammad Wasif Saif
- a Department of Hematology and Oncology , Tufts University School of Medicine , Boston , MA , USA
| |
Collapse
|
30
|
Andrikou K, Peterle C, Pipitone S, Salati M, Cascinu S. Emerging antibodies for the treatment of pancreatic cancer. Expert Opin Emerg Drugs 2017; 22:39-51. [DOI: 10.1080/14728214.2017.1293649] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Kalliopi Andrikou
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Chiara Peterle
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Stefania Pipitone
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Massimiliano Salati
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Stefano Cascinu
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| |
Collapse
|
31
|
Nowara E, Huszno J. Masitinib plus gemcitabine for personalized treatment of PDAC patients with overexpression of ACOX1. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016. [DOI: 10.1080/23808993.2016.1257911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Elzbieta Nowara
- Clinical and Experimental Chemotherapy Department, Cancer Center and Institution of Oncology, Gliwice, Poland
| | - Joanna Huszno
- Clinical and Experimental Chemotherapy Department, Cancer Center and Institution of Oncology, Gliwice, Poland
| |
Collapse
|
32
|
Karanikas M, Esempidis A, Chasan ZTM, Deftereou T, Antonopoulou M, Bozali F, Amarantidis K, Man YG. Pancreatic Cancer from Molecular Pathways to Treatment Opinion. J Cancer 2016; 7:1328-39. [PMID: 27390608 PMCID: PMC4934041 DOI: 10.7150/jca.15419] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/01/2016] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer is considered one of the most lethal malignances. It has been observed that the five year survival rate is less than 5%. Early diagnosis, understanding the risk factors and investigation of the molecular pathways with targeted therapy are the keys for efficient treatment. Moreover; there are several local treatments for patients with unresectable pancreatic cancer. There are several combined therapies with chemotherapy and radiotherapy, however; a local therapy approach for many patients with poor performance status are in need. For those patients with good performance status new polychemotherapy regimens are used with success and increased survival improvement. Polychemotherapy has been observed to increase the rate of radical resections in some cases. Second line therapy is used for patients with good performance status and metastatic disease. Oxaliplatin-based regimens are mostly used, however; there are several other drugs that are being developed. Unfortunately, targeted therapy has not presented the expected efficiency. Moreover; immunotherapy; another treatment approach for several cancers types has again failed to present positive results for pancreatic cancer. In the current mini review, we will present information from the diagnosis to molecular pathways and targeted treatment.
Collapse
Affiliation(s)
- Michail Karanikas
- 1. 1ST Department of Surgery, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, Alexandroupolis, 68100 Thrace, Greece
| | - Agis Esempidis
- 1. 1ST Department of Surgery, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, Alexandroupolis, 68100 Thrace, Greece
| | - Zeinep Tzoutze Memet Chasan
- 1. 1ST Department of Surgery, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, Alexandroupolis, 68100 Thrace, Greece
| | - Theodora Deftereou
- 1. 1ST Department of Surgery, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, Alexandroupolis, 68100 Thrace, Greece
| | - Maria Antonopoulou
- 1. 1ST Department of Surgery, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, Alexandroupolis, 68100 Thrace, Greece
| | - Ferdi Bozali
- 1. 1ST Department of Surgery, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, Alexandroupolis, 68100 Thrace, Greece
| | - Kyriakos Amarantidis
- 2. Department of Medical Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, Alexandroupolis, 68100 Thrace, Greece
| | - Yan-Gao Man
- 3. Research Laboratory and International Collaboration, Bon Secours Cancer Institute, VA, USA
| |
Collapse
|
33
|
Kosmidis C, Sapalidis K, Kotidis E, Mixalopoulos N, Zarogoulidis P, Tsavlis D, Baka S, Man YG, Kanellos J. Pancreatic cancer from bench to bedside: molecular pathways and treatment options. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:165. [PMID: 27275478 PMCID: PMC4876273 DOI: 10.21037/atm.2016.05.11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 03/24/2016] [Indexed: 12/15/2022]
Abstract
In the last forty years the pancreatic cancer treatment has made advances, however; still novel drugs are needed. It is known that the five year survival rate remains around 5%. The best treatment option still remains surgery, if patients are diagnosed early. In the last decade the biology of pancreatic cancer has been vastly explored and novel agents such as; tyrosine kinase agents, or vaccines have been added as a treatment perspective. The big challenge is now to translate this knowledge in better outcomes for patients. In this current review we will present information from pancreatic cancer diagnosis to molecular pathways and treatment options; current and future.
Collapse
|
34
|
Elebro J, Heby M, Warfvinge CF, Nodin B, Eberhard J, Jirström K. Expression and Prognostic Significance of Human Epidermal Growth Factor Receptors 1, 2 and 3 in Periampullary Adenocarcinoma. PLoS One 2016; 11:e0153533. [PMID: 27070783 PMCID: PMC4829175 DOI: 10.1371/journal.pone.0153533] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 03/30/2016] [Indexed: 12/11/2022] Open
Abstract
Periampullary adenocarcinoma, including pancreatic cancer, is a heterogeneous group of tumours with dismal prognosis, for which there is an urgent need to identify novel treatment strategies. The human epithelial growth factor receptors EGFR, HER2 and HER3 have been studied in several tumour types, and HER-targeting drugs have a beneficial effect on survival in selected types of cancer. However, these effects have not been evident in pancreatic cancer, and remain unexplored in other types of periampullary cancer. The prognostic impact of HER-expression in these cancers also remains unclear. The aim of this study was therefore to examine the expression and prognostic value of EGFR, HER2 and HER3 in periampullary cancer, with particular reference to histological subtype. To this end, protein expression of EGFR, HER2 and HER3, and HER2 gene amplification was assessed by immunohistochemistry and silver in situ hybridization, respectively, on tissue microarrays with tumours from 175 periampullary adenocarcinomas, with follow-up data on recurrence-free survival (RFS) and overall survival (OS) for up to 5 years. EGFR expression was similar in pancreatobiliary (PB) and intestinal (I) type tumours, but high HER2 and HER3 expression was significantly more common in I-type tumours. In PB-type cases receiving adjuvant gemcitabine, but not in untreated cases, high EGFR expression was significantly associated with a shorter OS and RFS, with a significant treatment interaction in relation to OS (pinteraction = 0.042). In I-type cases, high EGFR expression was associated with a shorter OS and RFS in univariable, but not in multivariable, analysis. High HER3 expression was associated with a prolonged RFS in univariable, but not in multivariable, analysis. Neither HER2 protein expression nor gene amplification was prognostic. The finding of a potential interaction between the expression of EGFR and response to adjuvant chemotherapy in PB-type tumours needs validation, and merits further study.
Collapse
Affiliation(s)
- Jacob Elebro
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Skåne University Hospital, 221 85, Lund, Sweden
- * E-mail:
| | - Margareta Heby
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Skåne University Hospital, 221 85, Lund, Sweden
| | - Carl Fredrik Warfvinge
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Skåne University Hospital, 221 85, Lund, Sweden
| | - Björn Nodin
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Skåne University Hospital, 221 85, Lund, Sweden
| | - Jakob Eberhard
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Skåne University Hospital, 221 85, Lund, Sweden
| | - Karin Jirström
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Skåne University Hospital, 221 85, Lund, Sweden
| |
Collapse
|
35
|
Assenat E, Azria D, Mollevi C, Guimbaud R, Tubiana-Mathieu N, Smith D, Delord JP, Samalin E, Portales F, Larbouret C, Robert B, Bibeau F, Bleuse JP, Crapez E, Ychou M, Pèlegrin A. Dual targeting of HER1/EGFR and HER2 with cetuximab and trastuzumab in patients with metastatic pancreatic cancer after gemcitabine failure: results of the "THERAPY"phase 1-2 trial. Oncotarget 2016; 6:12796-808. [PMID: 25918250 PMCID: PMC4494975 DOI: 10.18632/oncotarget.3473] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/22/2015] [Indexed: 12/12/2022] Open
Abstract
To improve treatment efficacy, we decided to simultaneously target HER1 and HER2 with trastuzumab and cetuximab. Following promising preclinical results, we conducted a phase 1-2 trial in advanced pancreatic cancer patients after first-line gemcitabine-based chemotherapy failure. In this single-arm, non-randomized, multicenter trial, patients received weekly cetuximab (400mg/m², then 250mg/m²). They were sequentially included in two trastuzumab dose levels: 3.0 or 4.0mg/kg, then 1.5 or 2.0mg/kg/weekly. Endpoints were the objective response rate, safety, progression-free (PFS) and overall survival (OS). During phase 1 (n=10 patients), toxicities were evenly distributed except for skin toxicities that frequently caused compliance issues. The higher dose level was defined as the trastuzumab recommended dose. During phase 2 (n=39 patients), toxicities were mainly cutaneous reactions and asthenia. No objective response was observed. Nine patients were stabilized but arrested treatment due to toxicity. Median PFS was 1.8 months (95%CI: 1.7-2.0 months) and median OS was 4.6 months (95%CI: 2.7–6.6 months). Both were positively correlated with skin toxicity severity (P=0.027 and P=0.001, respectively). Conventional phase 1 dose-escalation schedules are unsuitable for targeted therapies because most cutaneous toxicities are not considered dose-limiting toxicities. The compliance issues caused by skin toxicities were particularly detrimental because of the toxicity-response correlation.
Collapse
Affiliation(s)
- Eric Assenat
- Centre Hospitalier Régional Universitaire (CHU) de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - David Azria
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France.,IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U896, Montpellier, France.,Université Montpellier 1, Montpellier, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, France
| | - Caroline Mollevi
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Rosine Guimbaud
- Centre Hospitalier Universitaire (CHU) de Toulouse, TSA, Toulouse Cedex, France
| | | | - Denis Smith
- Centre Hospitalier Universitaire (CHU) de Bordeaux, Talence Cedex, France
| | | | - Emmanuelle Samalin
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Fabienne Portales
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Christel Larbouret
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U896, Montpellier, France.,Université Montpellier 1, Montpellier, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, France
| | - Bruno Robert
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U896, Montpellier, France.,Université Montpellier 1, Montpellier, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, France
| | - Frédéric Bibeau
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Jean-Pierre Bleuse
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Evelyne Crapez
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Marc Ychou
- Centre Hospitalier Régional Universitaire (CHU) de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France.,IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U896, Montpellier, France.,Université Montpellier 1, Montpellier, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, France
| | - André Pèlegrin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM, U896, Montpellier, France.,Université Montpellier 1, Montpellier, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, France
| |
Collapse
|
36
|
Shultz DB, Pai J, Chiu W, Ng K, Hellendag MG, Heestand G, Chang DT, Tu D, Moore MJ, Parulekar WR, Koong AC. A Novel Biomarker Panel Examining Response to Gemcitabine with or without Erlotinib for Pancreatic Cancer Therapy in NCIC Clinical Trials Group PA.3. PLoS One 2016; 11:e0147995. [PMID: 26808546 PMCID: PMC4725948 DOI: 10.1371/journal.pone.0147995] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/10/2016] [Indexed: 12/12/2022] Open
Abstract
Purpose NCIC Clinical Trials Group PA.3 was a randomized control trial that demonstrated improved overall survival (OS) in patients receiving erlotinib in addition to gemcitabine for locally advanced or metastatic pancreatic cancer. Prior to therapy, patients had plasma samples drawn for future study. We sought to identify biomarkers within these samples. Experimental Design Using the proximity ligation assay (PLA), a probe panel was built from commercially available antibodies for 35 key proteins selected from a global genetic analysis of pancreatic cancers, and used to quantify protein levels in 20 uL of patient plasma. To determine if any of these proteins levels independently associated with OS, univariate and mulitbaraible Cox models were used. In addition, we examined the associations between biomarker expression and disease stage at diagnosis using Fisher's exact test. The correlation between Erlotinib sensitivity and each biomarkers was assessed using a test of interaction between treatment and biomarker. Results and Conclusion Of the 569 eligible patients, 480 had samples available for study. Samples were randomly allocated into training (251) and validation sets (229). Among all patients, elevated levels of interleukin-8 (IL-8), carcinoembryonic antigen (CEA), hypoxia-inducible factor 1-alpha (HIF-1 alpha), and interleukin-6 were independently associated with lower OS, while IL-8, CEA, platelet-derived growth factor receptor alpha and mucin-1 were associated with metastatic disease. Patients with elevated levels of receptor tyrosine-protein kinase erbB-2 (HER2) expression had improved OS when treated with erlotinib compared to placebo. In conclusion, PLA is a powerful tool for identifying biomarkers from archived, small volume serum samples. These data may be useful to stratify patient outcomes regardless of therapeutic intervention. Trial Registration ClinicalTrials.gov NCT00040183
Collapse
Affiliation(s)
| | - Jonathan Pai
- School of Medicine, University of California San Francisco, San Francisco, United States of America
| | - Wayland Chiu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Kendall Ng
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States of America
| | | | - Gregory Heestand
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States of America
| | - Daniel T. Chang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Dongsheng Tu
- NCIC Clinical Trials Group, Queen's University, Kingston, Canada
| | - Malcolm J. Moore
- British Columbia Cancer Agency, Vancouver, British Columbia, CA, United States of America
| | | | - Albert C. Koong
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States of America
- * E-mail:
| |
Collapse
|
37
|
Matsuoka T, Yashiro M. Molecular targets for the treatment of pancreatic cancer: Clinical and experimental studies. World J Gastroenterol 2016; 22:776-789. [PMID: 26811624 PMCID: PMC4716076 DOI: 10.3748/wjg.v22.i2.776] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/13/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is the fourth most common cause of cancer deaths worldwide. Although recent therapeutic developments for patients with pancreatic cancer have provided survival benefits, the outcomes for patients with pancreatic cancer remain unsatisfactory. Molecularly targeted cancer therapy has advanced in the past decade with the use of a number of pathways as candidates of therapeutic targets. This review summarizes the molecular features of this refractory disease while focusing on the recent clinical and experimental findings on pancreatic cancer. It also discusses the data supporting current standard clinical outcomes, and offers conclusions that may improve the management of pancreatic cancer in the future.
Collapse
|
38
|
Grimont A, Pinho AV, Cowley MJ, Augereau C, Mawson A, Giry-Laterrière M, Van den Steen G, Waddell N, Pajic M, Sempoux C, Wu J, Grimmond SM, Biankin AV, Lemaigre FP, Rooman I, Jacquemin P. SOX9 regulates ERBB signalling in pancreatic cancer development. Gut 2015; 64:1790-9. [PMID: 25336113 DOI: 10.1136/gutjnl-2014-307075] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 10/01/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The transcription factor SOX9 was recently shown to stimulate ductal gene expression in pancreatic acinar-to-ductal metaplasia and to accelerate development of premalignant lesions preceding pancreatic ductal adenocarcinoma (PDAC). Here, we investigate how SOX9 operates in pancreatic tumourigenesis. DESIGN We analysed genomic and transcriptomic data from surgically resected PDAC and extended the expression analysis to xenografts from PDAC samples and to PDAC cell lines. SOX9 expression was manipulated in human cell lines and mouse models developing PDAC. RESULTS We found genetic aberrations in the SOX9 gene in about 15% of patient tumours. Most PDAC samples strongly express SOX9 protein, and SOX9 levels are higher in classical PDAC. This tumour subtype is associated with better patient outcome, and cell lines of this subtype respond to therapy targeting epidermal growth factor receptor (EGFR/ERBB1) signalling, a pathway essential for pancreatic tumourigenesis. In human PDAC, high expression of SOX9 correlates with expression of genes belonging to the ERBB pathway. In particular, ERBB2 expression in PDAC cell lines is stimulated by SOX9. Inactivating Sox9 expression in mice confirmed its role in PDAC initiation; it demonstrated that Sox9 stimulates expression of several members of the ERBB pathway and is required for ERBB signalling activity. CONCLUSIONS By integrating data from patient samples and mouse models, we found that SOX9 regulates the ERBB pathway throughout pancreatic tumourigenesis. Our work opens perspectives for therapy targeting tumourigenic mechanisms.
Collapse
Affiliation(s)
- Adrien Grimont
- Université catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Andreia V Pinho
- Cancer Research Division, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia Australian Pancreatic Cancer Genome Initiative
| | - Mark J Cowley
- Cancer Research Division, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia Australian Pancreatic Cancer Genome Initiative
| | - Cécile Augereau
- Université catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Amanda Mawson
- Cancer Research Division, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia Australian Pancreatic Cancer Genome Initiative
| | - Marc Giry-Laterrière
- Cancer Research Division, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia Australian Pancreatic Cancer Genome Initiative
| | | | - Nicola Waddell
- Australian Pancreatic Cancer Genome Initiative Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Marina Pajic
- Cancer Research Division, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia Australian Pancreatic Cancer Genome Initiative St Vincent's Clinical School, University New South Wales, Australia
| | - Christine Sempoux
- Department of Pathology, Université catholique de Louvain, Cliniques Universitaires St Luc, Brussels, Belgium
| | - Jianmin Wu
- Cancer Research Division, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia Australian Pancreatic Cancer Genome Initiative St Vincent's Clinical School, University New South Wales, Australia
| | - Sean M Grimmond
- Australian Pancreatic Cancer Genome Initiative Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia Wolfson Wohl Cancer Centre, University of Glasgow, Scotland, UK
| | - Andrew V Biankin
- Cancer Research Division, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia Australian Pancreatic Cancer Genome Initiative St Vincent's Clinical School, University New South Wales, Australia Wolfson Wohl Cancer Centre, University of Glasgow, Scotland, UK
| | | | - Ilse Rooman
- Cancer Research Division, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia Australian Pancreatic Cancer Genome Initiative St Vincent's Clinical School, University New South Wales, Australia
| | - Patrick Jacquemin
- Université catholique de Louvain, de Duve Institute, Brussels, Belgium
| |
Collapse
|
39
|
Kan S, Koido S, Okamoto M, Hayashi K, Ito M, Kamata Y, Komita H, Nagasaki E, Homma S. Up-regulation of HER2 by gemcitabine enhances the antitumor effect of combined gemcitabine and trastuzumab emtansine treatment on pancreatic ductal adenocarcinoma cells. BMC Cancer 2015; 15:726. [PMID: 26475267 PMCID: PMC4609140 DOI: 10.1186/s12885-015-1772-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 10/10/2015] [Indexed: 02/06/2023] Open
Abstract
Background Although pancreatic ductal adenocarcinomas (PDAs) widely express HER2, the expression level is generally low. If HER2 expression in PDA cells could be enhanced by treatment with a given agent, then combination therapy with that agent and trastuzumab emtansine (T-DM1), a chemotherapeutic agent that is a conjugate of trastuzumab, might lead to significant antitumor effects against PDA. Methods Cell proliferation was examined by spectrophotometry. HER2 expression was examined by flow cytometry, immunoblot and quantitative reverse transcription polymerase chain reaction. T-DM1 binding to cells was examined by flow cytometry and enzyme-linked immunosorbent assay. Results Out of 5 tested human PDA cell lines, including MIA PaCa-2, three showed increases in HER2 expression after gemcitabine (GEM) treatment. The binding of T-DM1 to GEM-treated MIA PaCa-2 cells was higher than to untreated MIA PaCa-2 cells. Treatment with GEM and T-DM1 showed synergic cytotoxic effects on MIA PaCa-2 cells in vitro. Cells in the G2M phase of the cell cycle were retained after GEM treatment and showed higher levels of HER2 expression, possibly contributing to the synergic effect of GEM and T-DM1. Conclusions Combined treatment with GEM and T-DM1 might confer a potent therapeutic modality against PDA as a result of GEM-mediated HER2 up-regulation. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1772-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shin Kan
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan.
| | - Shigeo Koido
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan. .,Division of Gastroenter ology and Hepatology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan.
| | - Masato Okamoto
- Department of Advanced Immunotherapeutics, Kitasato University School of Pharmacy, Tokyo, Japan.
| | - Kazumi Hayashi
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan. .,Division of Oncology and Hematology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan.
| | - Masaki Ito
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan.
| | - Yuko Kamata
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan.
| | - Hideo Komita
- Shimbashi Medical Checkup Office, Jikei University Hospital, Tokyo, Japan.
| | - Eijiro Nagasaki
- Division of Oncology and Hematology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan.
| | - Sadamu Homma
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
40
|
Abstract
There is now compelling evidence that the molecular heterogeneity of cancer is associated with disparate phenotypes with variable outcomes and therapeutic responsiveness to therapy in histologically indistinguishable cancers. This diversity may explain why conventional clinical trial designs have mostly failed to show efficacy when patients are enrolled in an unselected fashion. Knowledge of the molecular phenotype has the potential to improve therapeutic selection and hence the early delivery of the optimal therapeutic regimen. Resolution of the challenges associated with a more stratified approach to health care will ensure more precise diagnostics and enhance therapeutic selection, which will improve overall outcomes.
Collapse
Affiliation(s)
- Nigel B Jamieson
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; Academic Unit of Surgery, School of Medicine, College of Medical, Veterinary and Life Sciences, Glasgow Royal Infirmary, University of Glasgow, Alexandra Parade, Glasgow G31 2ER, UK; West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Alexandra Parade, Glasgow G31 2ER, UK
| | - David K Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 370 Victoria Street, Darlinghurst, New South Wales 2010, Australia; Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia; Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, New South Wales 2200, Australia; Faculty of Medicine, South Western Sydney Clinical School, University of NSW, Goulburn St, Liverpool, New South Wales 2170, Australia
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 370 Victoria Street, Darlinghurst, New South Wales 2010, Australia; Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia; Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, New South Wales 2200, Australia; Faculty of Medicine, South Western Sydney Clinical School, University of NSW, Goulburn St, Liverpool, New South Wales 2170, Australia.
| |
Collapse
|
41
|
Omar N, Yan B, Salto-Tellez M. HER2: An emerging biomarker in non-breast and non-gastric cancers. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.pathog.2015.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
42
|
Chiorean EG, Coveler AL. Pancreatic cancer: optimizing treatment options, new, and emerging targeted therapies. Drug Des Devel Ther 2015; 9:3529-45. [PMID: 26185420 PMCID: PMC4500614 DOI: 10.2147/dddt.s60328] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer death in the US and is expected to become the second leading cause of cancer-related deaths in the next decade. Despite 5-fluorouracil/leucovorin with irinotecan and oxaliplatin (FOLFIRINOX) and gemcitabine/nab-paclitaxel significantly improving outcomes for metastatic cancer, refractory disease still poses significant challenges. Difficulties with early detection and the inherent chemo- and radio-resistant nature of this malignancy led to attempts to define the sequential biology of pancreatic cancer in order to improve survival outcomes. Pancreatic adenocarcinoma is characterized by several germline or acquired genetic mutations, the most common being KRAS (90%), CDK2NA (90%), TP53 (75%-90%), DPC4/SMAD4 (50%). In addition, the tumor microenvironment, chemoresistant cancer stem cells, and the desmoplastic stroma have been the target of some promising clinical investigations. Among the core pathways reproducibly shown to lead the development and progression of this disease, DNA repair, apoptosis, G1/S cell cycle transition, KRAS, Wnt, Notch, Hedgehog, TGF-beta, and other cell invasion pathways, have been the target of "precision therapeutics". No single molecularly targeted therapeutic though has been uniformly successful, probably due to the tumor heterogeneity, but biomarker research is evolving and it hopes to select more patients likely to benefit. Recent reports note activity with immunotherapies such as CD40 agonists, CCR2 inhibitors, cancer vaccines, and novel combinations against the immunosuppressive tumor milieu are ongoing. While many obstacles still exist, clearly we are making progress in deciphering the heterogeneity within pancreatic cancers. Integrating conventional and immunological targeting will be the key to effective treatment of this deadly disease.
Collapse
Affiliation(s)
| | - Andrew L Coveler
- Department of Medicine, Division of Oncology, University of Washington, Seattle, WA, USA
| |
Collapse
|
43
|
Seicean A, Petrusel L, Seicean R. New targeted therapies in pancreatic cancer. World J Gastroenterol 2015; 21:6127-6145. [PMID: 26034349 PMCID: PMC4445091 DOI: 10.3748/wjg.v21.i20.6127] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/26/2015] [Accepted: 04/16/2015] [Indexed: 02/06/2023] Open
Abstract
Patients with pancreatic cancer have a poor prognosis with a median survival of 4-6 mo and a 5-year survival of less than 5%. Despite therapy with gemcitabine, patient survival does not exceed 6 mo, likely due to natural resistance to gemcitabine. Therefore, it is hoped that more favorable results can be obtained by using guided immunotherapy against molecular targets. This review summarizes the new leading targeted therapies in pancreatic cancers, focusing on passive and specific immunotherapies. Passive immunotherapy may have a role for treatment in combination with radiochemotherapy, which otherwise destroys the immune system along with tumor cells. It includes mainly therapies targeting against kinases, including epidermal growth factor receptor, Ras/Raf/mitogen-activated protein kinase cascade, human epidermal growth factor receptor 2, insulin growth factor-1 receptor, phosphoinositide 3-kinase/Akt/mTOR and hepatocyte growth factor receptor. Therapies against DNA repair genes, histone deacetylases, microRNA, and pancreatic tumor tissue stromal elements (stromal extracellular matric and stromal pathways) are also discussed. Specific immunotherapies, such as vaccines (whole cell recombinant, peptide, and dendritic cell vaccines), adoptive cell therapy and immunotherapy targeting tumor stem cells, have the role of activating antitumor immune responses. In the future, treatments will likely include personalized medicine, tailored for numerous molecular therapeutic targets of multiple pathogenetic pathways.
Collapse
|
44
|
Goel G, Sun W. Novel approaches in the management of pancreatic ductal adenocarcinoma: potential promises for the future. J Hematol Oncol 2015; 8:44. [PMID: 25935754 PMCID: PMC4431030 DOI: 10.1186/s13045-015-0141-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 04/21/2015] [Indexed: 02/08/2023] Open
Abstract
Despite a few breakthroughs in therapy for advanced disease in the recent years, pancreatic ductal adenocarcinoma continues to remain one of the most challenging human malignancies to treat. The overall prognosis for the majority of patients with pancreatic cancer is rather dismal, and therefore, more effective treatment options are being desperately sought. The practical goals of management are to improve the cure rates for patients with resectable disease, achieve a higher conversion rate of locally advanced tumor into potentially resectable disease, and finally, prolong the overall survival for those who develop metastatic disease. Our understanding of the complex genetic alterations, the implicated molecular pathways, and the role of desmoplastic stroma in pancreatic cancer tumorigenesis has increased several folds in the recent years. This has facilitated the development of novel therapeutic strategies against pancreatic cancer, some of which are currently under evaluation in ongoing preclinical and clinical studies. This review will summarize the existing treatment approaches for this devastating disease and also discuss the promising therapeutic approaches that are currently in different stages of clinical development.
Collapse
Affiliation(s)
- Gaurav Goel
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, 5150 Centre Avenue, Fifth Floor, Pittsburgh, PA, 15232, USA.
| | - Weijing Sun
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, 5150 Centre Avenue, Fifth Floor, Pittsburgh, PA, 15232, USA.
| |
Collapse
|
45
|
Abstract
Pancreatic cancer is expected to be the second deadliest malignancy in the USA by 2020. The survival rates for patients with other gastrointestinal malignancies have increased consistently during the past 30 years; unfortunately, however, the outcomes of patients with pancreatic cancer have not changed significantly. Although surgery remains the only curative treatment for pancreatic cancer, therapeutic strategies based on initial resection have not substantially improved the survival of patients with resectable disease over the past 25 years; presently, more than 80% of patients suffer disease relapse after resection. Preclinical evidence that pancreatic cancer is a systemic disease suggests a possible benefit for early administration of systemic therapy in these patients. In locally advanced disease, the role of chemoradiotherapy is increasingly being questioned, particularly considering the results of the LAP-07 trial. Novel biomarkers are clearly needed to identify subsets of patients likely to benefit from chemoradiotherapy. In the metastatic setting, FOLFIRINOX (folinic acid, 5-fluorouracil, irinotecan, and oxaliplatin), and nab-paclitaxel plus gemcitabine have yielded only modest improvements in survival. Thus, new treatments are urgently needed for patients with pancreatic cancer. Herein, we review the state-of-the-art of pancreatic cancer treatment, and the upcoming novel therapeutics that hold promise in this disease are also discussed.
Collapse
|
46
|
CHO ILRAE, KAOWINN SIRICHAT, MOON JEONG, SOH JIWON, KANG HOYOUNG, JUNG CHOROK, OH SANGTAEK, SONG HAYNE, KOH SANGSEOK, CHUNG YOUNGHWA. Oncotropic H-1 parvovirus infection degrades HIF-1α protein in human pancreatic cancer cells independently of VHL and RACK1. Int J Oncol 2015; 46:2076-82. [DOI: 10.3892/ijo.2015.2922] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 12/05/2014] [Indexed: 11/06/2022] Open
|
47
|
A phase I trial of combination trastuzumab, lapatinib, and bevacizumab in patients with advanced cancer. Invest New Drugs 2014; 33:177-86. [PMID: 25323060 DOI: 10.1007/s10637-014-0173-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE Preclinical data indicate that combination HER2-directed and anti-VEGF therapy may bypass resistance to trastuzumab. A phase I trial was performed to assess safety, activity, and correlates. EXPERIMENTAL DESIGN Patients with advanced, refractory malignancy were enrolled (modified 3 + 3 design with expansions for responding tumor types). Patients received lapatinib daily for 21 days, and bevacizumab and trastuzumab every 3 weeks. Correlates included HER2 extracellular domain levels (ECD) and single nucleotide polymorphisms (SNPs). RESULTS Ninety-four patients were treated (median = four prior systemic therapies). The most common related adverse events ≥ grade 2 were diarrhea (n = 33, 35 %) and hypertension (n = 10, 11 %). The recommended phase 2 dose was trastuzumab 6 mg/m(2) (loading = 8 mg/m(2)) and bevacizumab 15 mg/kg every 3 weeks, with lapatinib 1,250 mg daily (full FDA-approved dose of each drug). One patient (1 %) achieved a complete response (CR); eight (9 %), a partial response (PR) (includes breast (n = 7, one of which was HER2 2+ by IHC) and salivary ductal carcinoma (n = 1); and 14 (15 %), stable disease (SD) ≥6 months (total SD ≥ 6 months/PR/CR =23 (25 %). All patients with PR/CR received prior trastuzumab +/- lapatinib. SD ≥ 6 months/PR/CR rate and time to treatment failure (TTF) correlated with elevated baseline HER2 ECD (N = 75 patients tested) but not with HER2 SNPs. CONCLUSIONS Combination trastuzumab, lapatinib, and bevacizumab was well-tolerated and demonstrated antitumor activity in heavily pretreated patients with advanced malignancy.
Collapse
|
48
|
Chiu JW, Wong H, Leung R, Pang R, Cheung TT, Fan ST, Poon R, Yau T. Advanced pancreatic cancer: flourishing novel approaches in the era of biological therapy. Oncologist 2014; 19:937-50. [PMID: 25117068 PMCID: PMC4153449 DOI: 10.1634/theoncologist.2012-0131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 06/06/2014] [Indexed: 12/13/2022] Open
Abstract
The progress in the development of systemic treatment for advanced pancreatic cancer (APC) has been slow. The mainstream treatment remains using chemotherapy including gemcitabine, FOLFIRINOX, and nab-paclitaxel. Erlotinib is the only approved biological therapy with marginal benefit. Studies of agents targeting epidermal growth factor receptor, angiogenesis, and RAS signaling have not been satisfying, and the usefulness of targeted therapy in APC is uncertain. Understanding in molecular processes and tumor biology has opened the door for new treatment strategies such as targeting insulin-like growth factor 1 receptor, transforming growth factor β, phosphoinositide 3-kinase/AKT/mammalian target of rapamycin pathway, and Notch pathway. New directions also include the upcoming immunotherapy and many novel agents that act on the microenvironment. The practice of personalized medicine using predictive biomarkers and pharmacogenomics signatures may also enhance the effectiveness of existing treatment. Future treatment approaches may involve comprehensive genomic assessment of tumor and integrated combinations of multiple agents to overcome treatment resistance.
Collapse
Affiliation(s)
- Joanne W Chiu
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Hilda Wong
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Roland Leung
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Roberta Pang
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Tan-To Cheung
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Sheung-Tat Fan
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Ronnie Poon
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Thomas Yau
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
49
|
Dual targeting of ErbB-2/ErbB-3 results in enhanced antitumor activity in preclinical models of pancreatic cancer. Oncogenesis 2014; 3:e117. [PMID: 25133484 PMCID: PMC5189962 DOI: 10.1038/oncsis.2014.31] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/10/2014] [Accepted: 07/15/2014] [Indexed: 12/16/2022] Open
Abstract
ErbB-3 and its ligand NRG-1β are key players in driving oncogenic signaling and resistance to therapy through the activation of the PI3K/Akt pathway. We have recently reported that EV20, a humanized anti-ErbB3 antibody, possesses a marked antitumor activity in a variety of human tumor models, including pancreatic cancer (PC). Here, we report that despite epidermal growth factor receptor overexpression, PC cells are more sensitive to NRG-1β than EGF in terms of Akt activation and cell proliferation. Using stable ErbB-3-knocked down cells and EV20 in combination with trastuzumab, we showed that dual targeting of ErbB-2 and ErbB-3 was necessary to completely abrogate ErbB-3 signaling and to impair cell proliferation. Similarly, enhanced therapeutic efficacy of the antibody combination was seen in xenografts originating from K-Ras-mutated HPAF-II and SW1990 cells, without increasing the toxicity. These results indicate that dual targeting of ErbB-2 and ErbB-3 could represent a new therapeutic approach in PC.Oncogenesis (2014) 3, e117; doi:10.1038/oncsis.2014.31; published online 18 August 2014.
Collapse
|
50
|
Chuong MD, Boggs DH, Patel KN, Regine WF. Adjuvant chemoradiation for pancreatic cancer: what does the evidence tell us? J Gastrointest Oncol 2014; 5:166-77. [PMID: 24982765 DOI: 10.3978/j.issn.2078-6891.2014.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/08/2014] [Indexed: 12/12/2022] Open
Abstract
The role of adjuvant chemoradiation (CRT) for pancreas cancer remains unclear. A handful of randomized trials conducted decades of ago ignited a debate that continues today about whether CRT improves survival after surgery. The many flaws in these trials are well described in the literature, which include the use of antiquated radiation delivery techniques and suboptimal doses. Recent prospective randomized data is lacking, and we eagerly await the results the ongoing Radiation Therapy Oncology Group (RTOG) 0848 trial that is evaluating the utility of high quality adjuvant CRT in resected pancreas cancer patients. Until the results of RTOG 0848 are available we should look to other studies from the modern era to guide adjuvant treatment recommendations. Here we review the current state of the art for adjuvant pancreas CRT with respect to patient selection, radiation techniques, radiation dose, and integration with novel systemic agents.
Collapse
Affiliation(s)
- Michael D Chuong
- Department of Radiation Oncology, University of Maryland Medical Systems, Baltimore, MD 21201, USA
| | - Drexell H Boggs
- Department of Radiation Oncology, University of Maryland Medical Systems, Baltimore, MD 21201, USA
| | - Kruti N Patel
- Department of Radiation Oncology, University of Maryland Medical Systems, Baltimore, MD 21201, USA
| | - William F Regine
- Department of Radiation Oncology, University of Maryland Medical Systems, Baltimore, MD 21201, USA
| |
Collapse
|