51
|
Nakai Y, Takahara N, Mizuno S, Kogure H, Koike K. Current Status of Endoscopic Ultrasound Techniques for Pancreatic Neoplasms. Clin Endosc 2019; 52:527-532. [PMID: 31337194 PMCID: PMC6900295 DOI: 10.5946/ce.2019.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/13/2019] [Indexed: 12/15/2022] Open
Abstract
Endoscopic ultrasound (EUS) now plays an important role in the management of pancreatic neoplasms. There are various types of pancreatic neoplasms, from benign to malignant lesions, and the role of EUS ranges from the imaging diagnosis to treatment. EUS is useful for the detection, characterization, and tissue acquisition of pancreatic lesions. Recent advancement of contrast-enhanced harmonic EUS and elastography enables better characterization of pancreatic lesions. In addition to these enhanced EUS imaging techniques, EUS-guided tissue acquisition is now the standard procedure to establish the pathological diagnosis of pancreatic neoplasms. While these diagnostic roles of EUS have been established, EUS-guided interventions such as ablation and drainage are also increasingly utilized in the management of pancreatic neoplasms. However, most of these EUS-guided interventions are not yet standardized in terms of techniques and devices and thus need further investigations.
Collapse
Affiliation(s)
- Yousuke Nakai
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naminatsu Takahara
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Suguru Mizuno
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirofumi Kogure
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
52
|
Obesity-Induced Adipose Tissue Inflammation as a Strong Promotional Factor for Pancreatic Ductal Adenocarcinoma. Cells 2019; 8:cells8070673. [PMID: 31277269 PMCID: PMC6678863 DOI: 10.3390/cells8070673] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is expected to soon become the second leading cause of cancer related deaths in the United States. This may be due to the rising obesity prevalence, which is a recognized risk factor for PDAC. There is great interest in deciphering the underlying driving mechanisms of the obesity–PDAC link. Visceral adiposity has a strong correlation to certain metabolic diseases and gastrointestinal cancers, including PDAC. In fact, our own data strongly suggest that visceral adipose tissue inflammation is a strong promoter for PDAC growth and progression in a genetically engineered mouse model of PDAC and diet-induced obesity. In this review, we will discuss the relationship between obesity-associated adipose tissue inflammation and PDAC development, with a focus on the key molecular and cellular components in the dysfunctional visceral adipose tissue, which provides a tumor permissive environment.
Collapse
|
53
|
Palata O, Hradilova Podzimkova N, Nedvedova E, Umprecht A, Sadilkova L, Palova Jelinkova L, Spisek R, Adkins I. Radiotherapy in Combination With Cytokine Treatment. Front Oncol 2019; 9:367. [PMID: 31179236 PMCID: PMC6538686 DOI: 10.3389/fonc.2019.00367] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/23/2019] [Indexed: 12/22/2022] Open
Abstract
Radiotherapy (RT) plays an important role in the management of cancer patients. RT is used in more than 50% of patients during the course of their disease in a curative or palliative setting. In the past decades it became apparent that the abscopal effect induced by RT might be dependent on the activation of immune system, and that the induction of immunogenic cancer cell death and production of danger-associated molecular patterns from dying cells play a major role in the radiotherapy-mediated anti-tumor efficacy. Therefore, the combination of RT and immunotherapy is of a particular interest that is reflected in designing clinical trials to treat patients with various malignancies. The use of cytokines as immunoadjuvants in combination with RT has been explored over the last decades as one of the immunotherapeutic combinations to enhance the clinical response to anti-cancer treatment. Here we review mainly the data on the efficacy of IFN-α, IL-2, IL-2-based immunocytokines, GM-CSF, and TNF-α used in combinations with various radiotherapeutic techniques in clinical trials. Moreover, we discuss the potential of IL-15 and its analogs and IL-12 cytokines in combination with RT based on the efficacy in preclinical mouse tumor models.
Collapse
Affiliation(s)
- Ondrej Palata
- SOTIO a.s, Prague, Czechia.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| | - Nada Hradilova Podzimkova
- SOTIO a.s, Prague, Czechia.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| | | | | | | | - Lenka Palova Jelinkova
- SOTIO a.s, Prague, Czechia.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| | - Radek Spisek
- SOTIO a.s, Prague, Czechia.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| | - Irena Adkins
- SOTIO a.s, Prague, Czechia.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| |
Collapse
|
54
|
Aliru ML, Schoenhals JE, Venkatesulu BP, Anderson CC, Barsoumian HB, Younes AI, K Mahadevan LS, Soeung M, Aziz KE, Welsh JW, Krishnan S. Radiation therapy and immunotherapy: what is the optimal timing or sequencing? Immunotherapy 2019; 10:299-316. [PMID: 29421979 DOI: 10.2217/imt-2017-0082] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Radiotherapy is a component of the standard of care for many patients with locally advanced nonmetastatic tumors and increasingly those with oligometastatic tumors. Despite encouraging advances in local control and progression-free and overall survival outcomes, continued manifestation of tumor progression or recurrence leaves room for improvement in therapeutic efficacy. Novel combinations of radiation with immunotherapy have shown promise in improving outcomes and reducing recurrences by overcoming tumor immune tolerance and evasion mechanisms via boosting the immune system's ability to recognize and eradicate tumor cells. In this review, we discuss preclinical and early clinical evidence that radiotherapy and immunotherapy can improve treatment outcomes for locally advanced and metastatic tumors, elucidate underlying molecular mechanisms and address strategies to optimize timing and sequencing of combination therapy for maximal synergy.
Collapse
Affiliation(s)
- Maureen L Aliru
- From the Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.,Medical Physics Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Jonathan E Schoenhals
- From the Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Bhanu P Venkatesulu
- From the Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Clark C Anderson
- Departments of Internal Medicine & Molecular & Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
| | - Hampartsoum B Barsoumian
- From the Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Ahmed I Younes
- From the Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Lakshmi S K Mahadevan
- From the Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Melinda Soeung
- From the Departments of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kathryn E Aziz
- From the Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - James W Welsh
- From the Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.,From the Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sunil Krishnan
- From the Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.,From the Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Medical Physics Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
55
|
Nabi Z, Reddy DN. Endoscopic Palliation for Biliary and Pancreatic Malignancies: Recent Advances. Clin Endosc 2019; 52:226-234. [PMID: 30665289 PMCID: PMC6547342 DOI: 10.5946/ce.2019.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
Malignancies of the pancreatobiliary system are usually unresectable at the time of diagnosis. As a consequence, a majority of these cases are candidates for palliative care. With advances in chemotherapeutic agents and multidisciplinary care, the survival rate in pancreatobiliary malignancies has improved. Therefore, there is a need to provide an effective and long-lasting palliative care for these patients. Endoscopic palliation is preferred to surgery as the former is associated with equal efficacy and reduced morbidity. The main role of endoscopic palliation in the vast majority of pancreatobiliary malignancies includes biliary and enteral stenting for malignant obstructive jaundice and gastric outlet obstruction, respectively. Recent advances in endoscopic palliation appear promising in imparting long-lasting relief of symptoms. Use of radiofrequency ablation and photodynamic therapy in malignant biliary obstruction has been shown to improve the survival rates as well as the patency of biliary stents. The emergence of endoscopic ultrasound (EUS) as a therapeutic tool has enhanced the capability of minimally invasive palliation in pancreatobiliary cancers. EUS is a valuable alternative to endoscopic retrograde cholangiopancreatography for the palliation of obstructive jaundice. More recently, EUS is emerging as an effective primary modality for biliary and gastric bypass.
Collapse
Affiliation(s)
- Zaheer Nabi
- Asian Institute of Gastroenterology, Hyderabad, India
| | | |
Collapse
|
56
|
Boldrini L, Cusumano D, Cellini F, Azario L, Mattiucci GC, Valentini V. Online adaptive magnetic resonance guided radiotherapy for pancreatic cancer: state of the art, pearls and pitfalls. Radiat Oncol 2019; 14:71. [PMID: 31036034 PMCID: PMC6489212 DOI: 10.1186/s13014-019-1275-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 04/11/2019] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Different studies have proved in recent years that hypofractionated radiotherapy (RT) improves overall survival of patients affected by locally advanced, unresectable, pancreatic cancer. The clinical management of these patients generally leads to poor results and is considered very challenging, due to different factors, heavily influencing treatment delivery and its outcomes. Firstly, the dose prescribed to the target is limited by the toxicity that the highly radio-sensitive organs at risk (OARs) surrounding the disease can develop. Treatment delivery is also complicated by the significant inter-fractional and intra-fractional variability of therapy volumes, mainly related to the presence of hollow organs and to the breathing cycle. The recent introduction of magnetic resonance guided radiotherapy (MRgRT) systems leads to the opportunity to control most of the aforementioned sources of uncertainty influencing RT treatment workflow in pancreatic cancer. MRgRT offers the possibility to accurately identify radiotherapy volumes, thanks to the high soft-tissue contrast provided by the Magnetic Resonance imaging (MRI), and to monitor the tumour and OARs positions during the treatment fraction using a high-temporal cine MRI. However, the main advantage offered by the MRgRT is the possibility to online adapt the RT treatment plan, changing the dose distribution while the patient is still on couch and successfully addressing most of the sources of variability. SHORT CONCLUSION Aim of this study is to present and discuss the state of the art, the main pitfalls and the innovative opportunities offered by online adaptive MRgRT in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Luca Boldrini
- Dipartimento di Diagnostica per immagini, Radioterapia Oncologica ed Ematologia, UOC Radioterapia Oncologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Roma, Italia
| | - Davide Cusumano
- Dipartimento di Diagnostica per immagini, Radioterapia Oncologica ed Ematologia, UOC Fisica Sanitaria, Fondazione Policlinico Policlinico Universitario “A. Gemelli” IRCCS, Roma, Italia
| | - Francesco Cellini
- Dipartimento di Diagnostica per immagini, Radioterapia Oncologica ed Ematologia, UOC Radioterapia Oncologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Roma, Italia
| | - Luigi Azario
- Dipartimento di Diagnostica per immagini, Radioterapia Oncologica ed Ematologia, UOC Fisica Sanitaria, Fondazione Policlinico Policlinico Universitario “A. Gemelli” IRCCS, Roma, Italia
| | - Gian Carlo Mattiucci
- Dipartimento di Diagnostica per immagini, Radioterapia Oncologica ed Ematologia, UOC Radioterapia Oncologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Roma, Italia
| | - Vincenzo Valentini
- Dipartimento di Diagnostica per immagini, Radioterapia Oncologica ed Ematologia, UOC Radioterapia Oncologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Roma, Italia
| |
Collapse
|
57
|
Inflammation and Pancreatic Cancer: Focus on Metabolism, Cytokines, and Immunity. Int J Mol Sci 2019; 20:ijms20030676. [PMID: 30764482 PMCID: PMC6387440 DOI: 10.3390/ijms20030676] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/23/2019] [Accepted: 01/31/2019] [Indexed: 12/24/2022] Open
Abstract
Systemic and local chronic inflammation might enhance the risk of pancreatic ductal adenocarcinoma (PDAC), and PDAC-associated inflammatory infiltrate in the tumor microenvironment concurs in enhancing tumor growth and metastasis. Inflammation is closely correlated with immunity, the same immune cell populations contributing to both inflammation and immune response. In the PDAC microenvironment, the inflammatory cell infiltrate is unbalanced towards an immunosuppressive phenotype, with a prevalence of myeloid derived suppressor cells (MDSC), M2 polarized macrophages, and Treg, over M1 macrophages, dendritic cells, and effector CD4⁺ and CD8⁺ T lymphocytes. The dynamic and continuously evolving cross-talk between inflammatory and cancer cells might be direct and contact-dependent, but it is mainly mediated by soluble and exosomes-carried cytokines. Among these, tumor necrosis factor alpha (TNFα) plays a relevant role in enhancing cancer risk, cancer growth, and cancer-associated cachexia. In this review, we describe the inflammatory cell types, the cytokines, and the mechanisms underlying PDAC risk, growth, and progression, with particular attention on TNFα, also in the light of the potential risks or benefits associated with anti-TNFα treatments.
Collapse
|
58
|
Chang KJ. Endoscopic foregut surgery and interventions: The future is now. The state-of-the-art and my personal journey. World J Gastroenterol 2019; 25:1-41. [PMID: 30643356 PMCID: PMC6328959 DOI: 10.3748/wjg.v25.i1.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 02/06/2023] Open
Abstract
In this paper, I reviewed the emerging field of endoscopic surgery and present data supporting the contention that endoscopy can now be used to treat many foregut diseases that have been traditionally treated surgically. Within each topic, the content will progress as follows: "lessons learned", "technical considerations" and "future opportunities". Lessons learned will provide a brief background and update on the most current literature. Technical considerations will include my personal experience, including tips and tricks that I have learned over the years. Finally, future opportunities will address current unmet needs and potential new areas of development. The foregut is defined as "the upper part of the embryonic alimentary canal from which the pharynx, esophagus, lung, stomach, liver, pancreas, and part of the duodenum develop". Foregut surgery is well established in treating conditions such as gastroesophageal reflux disease (GERD), achalasia, esophageal diverticula, Barrett's esophagus (BE) and esophageal cancer, stomach cancer, gastric-outlet obstruction, and obesity. Over the past decade, remarkable progress in interventional endoscopy has culminated in the conceptualization and practice of endoscopic foregut surgery for various clinical conditions summarized in this paper. Regarding GERD, there are now several technologies available to effectively treat it and potentially eliminate symptoms, and the need for long-term treatment with proton pump inhibitors. For the first time, fundoplication can be performed without the need for open or laparoscopic surgery. Long-term data going out 5-10 years are now emerging showing extended durability. In respect to achalasia, per-oral endoscopic myotomy (POEM) which was developed in Japan, has become an alternative to the traditional Heller's myotomy. Recent meta-analysis show that POEM may have better results than Heller, but the issue of post-POEM GERD still needs to be addressed. There is now a resurgence of endoscopic treatment of Zenker's diverticula with improved technique (Z-POEM) and equipment; thus, patients are choosing flexible endoscopic treatment as opposed to open or rigid endoscopy options. In regard to BE, endoscopic submucosal dissection (ESD) which is well established in Asia, is now becoming more mainstream in the West for the treatment of BE with high grade dysplasia, as well as early esophageal cancer. In combination with all the ablation technologies (radiofrequency ablation, cryotherapy, hybrid argon plasma coagulation), the entire spectrum of Barrett's and related dysplasia and early cancer can be managed predominantly by endoscopy. Importantly, in regard to early gastric cancer and submucosal tumors (SMTs) of the stomach, ESD and full thickness resection (FTR) can excise these lesions en-bloc and endoscopic suturing is now used to close large defects and perforations. For treatment of patients with malignant gastric outlet obstruction (GOO), endoscopic gastro-jejunostomy is now showing better results than enteral stenting. G-POEM is also emerging as a treatment option for patients with gastroparesis. Obesity has become an epidemic in many western countries and is becoming also prevalent in Asia. Endoscopic sleeve gastroplasty (ESG) is now becoming an established treatment option, especially for obese patients with body mass index between 30 and 35. Data show an average weight loss of 16 kg after ESG with long-term data confirming sustainability. Finally, in respect to endo-hepatology, there are many new endoscopic interventions that have been developed for patients with liver disease. Endoscopic ultrasound (EUS)-guided liver biopsy and EUS-guided portal pressure measurement are exciting new frontiers for the endo-hepatologists.
Collapse
Affiliation(s)
- Kenneth J Chang
- H.H. Chao Comprehensive Digestive Disease Center, University of California, Irvine Medical Center, Orange, CA 92868, United States
| |
Collapse
|
59
|
|
60
|
Wang SJ, Haffty B. Radiotherapy as a New Player in Immuno-Oncology. Cancers (Basel) 2018; 10:cancers10120515. [PMID: 30558196 PMCID: PMC6315809 DOI: 10.3390/cancers10120515] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/14/2022] Open
Abstract
Recent development in radiation biology has revealed potent immunogenic properties of radiotherapy in cancer treatments. However, antitumor immune effects of radiotherapy are limited by the concomitant induction of radiation-dependent immunosuppressive effects. In the growing era of immunotherapy, combining radiotherapy with immunomodulating agents has demonstrated enhancement of radiation-induced antitumor immune activation that correlated with improved treatment outcomes. Yet, how to optimally deliver combination therapy regarding dose-fractionation and timing of radiotherapy is largely unknown. Future prospective testing to fine-tune this promising combination of radiotherapy and immunotherapy is warranted.
Collapse
Affiliation(s)
- Shang-Jui Wang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany St., New Brunswick, NJ 08901, USA.
| | - Bruce Haffty
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany St., New Brunswick, NJ 08901, USA.
| |
Collapse
|
61
|
Kamimura K, Yokoo T, Terai S. Gene Therapy for Pancreatic Diseases: Current Status. Int J Mol Sci 2018; 19:3415. [PMID: 30384450 PMCID: PMC6275054 DOI: 10.3390/ijms19113415] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/22/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022] Open
Abstract
The pancreas is a key organ involved in digestion and endocrine functions in the body. The major diseases of the pancreas include pancreatitis, pancreatic cancer, cystic diseases, pancreatic divisum, islet cell tumors, endocrine tumors, diabetes mellitus, and pancreatic pain induced by these diseases. While various therapeutic methodologies have been established to date, however, the improvement of conventional treatments and establishment of novel therapies are essential to improve the efficacy. For example, conventional therapeutic options, including chemotherapy, are not effective against pancreatic cancer, and despite improvements in the last decade, the mortality rate has not declined and is estimated to become the second cause of cancer-related deaths by 2030. Therefore, continuous efforts focus on the development of novel therapeutic options. In this review, we will summarize the progress toward the development of gene therapies for pancreatic diseases, with an emphasis on recent preclinical studies and clinical trials. We aim to identify new areas for improvement of the current methodologies and new strategies that will lead to safe and effective gene therapeutic approaches in pancreatic diseases.
Collapse
Affiliation(s)
- Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan.
| | - Takeshi Yokoo
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan.
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan.
| |
Collapse
|
62
|
Golan T, Geva R, Richards D, Madhusudan S, Lin BK, Wang HT, Walgren RA, Stemmer SM. LY2495655, an antimyostatin antibody, in pancreatic cancer: a randomized, phase 2 trial. J Cachexia Sarcopenia Muscle 2018; 9:871-879. [PMID: 30051975 PMCID: PMC6204586 DOI: 10.1002/jcsm.12331] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 06/10/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Cachexia is a formidable clinical challenge in pancreatic cancer. We assessed LY2495655 (antimyostatin antibody) plus standard-of-care chemotherapy in pancreatic cancer using cachexia status as a stratifier. METHODS In this randomized, phase 2 trial, patients with stage II-IV pancreatic cancer were randomized to 300 mg LY2495655, 100 mg LY2495655, or placebo, plus physician-choice chemotherapy from a prespecified list of standard-of-care regimens for first and later lines of care. Investigational treatment was continued during second-line treatment. The primary endpoint was overall survival. RESULTS Overall, 125 patients were randomized. In August 2014, 300 mg LY2495655 was terminated due to imbalance in death rates between the treatment arms; in January 2015, 100 mg LY2495655 treatment was terminated due to futility. LY2495655 did not improve overall survival: the hazard ratio was 1.70 (90% confidence interval, 1.1-2.7) for 300 mg vs. placebo and 1.3 (0.82-2.1) for 100 mg vs. placebo (recommended doses). Progression-free survival results were consistent with the overall survival results. A numerically higher hazard ratio was observed in patients with weight loss (WL) of ≥5% (cachexia) than with <5% WL within 6 months before randomization. Subgroup analyses for patients stratified by WL in the 6 months preceding enrollment suggested that functional responses to LY2495655 (either dose) may have been superior in patients with <5% WL vs. patients with ≥5% WL. Among possibly drug-related adverse events, fatigue, diarrhoea, and anorexia were more common in LY2495655-treated than in placebo-treated patients. CONCLUSIONS In the intention-to-treat analysis, LY2495655 did not confer clinical benefit in pancreatic cancer. Our data highlight the importance of assessing survival when investigating therapeutic management of cachexia and support the use of WL as a stratifier (independent of performance status).
Collapse
Affiliation(s)
| | - Ravit Geva
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Srinivasan Madhusudan
- Academic Oncology, University of Nottingham, School of Medicine, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| | | | | | | | - Salomon M Stemmer
- Rabin Medical Center, Petach Tikva, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
63
|
Evaluation of delivered dose to a moving target by 4D dose reconstruction in gated volumetric modulated arc therapy. PLoS One 2018; 13:e0202765. [PMID: 30192793 PMCID: PMC6128520 DOI: 10.1371/journal.pone.0202765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 08/08/2018] [Indexed: 12/11/2022] Open
Abstract
Purpose To develop a 4D dose reconstruction method and to evaluate the delivered dose in respiratory-gated volumetric modulated arc therapy (VMAT). Materials and methods A total 112 treatment sessions of gated VMAT for 30 stereotactic body radiotherapy (SBRT) patients (10 lung, 10 liver, and 10 pancreas) were evaluated. For respiratory-gated SBRT, 4DCT was acquired, and the CT data at the end-exhale phase was used for a VMAT plan. The delivered dose was reconstructed using a patient’s respiratory motion and machine motion acquired during the beam delivery. The machine motion was obtained from the treatment log file, while the target position was estimated from an external respiratory marker position. The target position was divided into 1-mm position bins, and sub-beams with beam isocenters corresponding to each position bin were created in a motion mimicking plan, reflecting motion data including MLC leaf positions and gantry angle and target position data during beam treatment. The reconstructed 4D dose was compared with the dose of the original plan using these dosimetric parameters; the maximum dose (Dmax) and mean dose (Dmean) of gross target volume (GTV) or organs at risk (spinal cord, esophagus, heart, duodenum, kidney, spinal cord, and stomach). The minimum dose (Dmin) to GTV was also calculated to verify cold spots in tumors. Results There was no significant difference of dose parameters regard to the GTV in all tumors. For the liver cases, there were significant differences in the Dmax of duodenum (-4.2 ± 1.4%), stomach (-3.5 ± 4.2%), left kidney (-4.1 ± 2.8%), and right kidney (-3.2 ± 1.3%), and in the Dmean of duodenum (-3.8 ± 1.4%), stomach (-3.9 ± 2.2%), left kidney (-3.1 ± 2.8%), and right kidney (-4.1 ± 2.6%). For the pancreas cases, there were significant differences in the Dmax of stomach (2.1 ± 3.0%), and in the Dmean of liver (1.5 ± 0.6%), duodenum (-1.0 ± 1.4%), stomach (2.1 ± 1.6%), and right kidney (-1.3 ± 0.9%). The average gamma pass rates were 97.6 ± 4.8% for lung cases, 99.6 ± 0.5% for liver cases, and 99.5 ± 0.5% for pancreas cases. Most cases showed insignificant dose variation, with gamma pass rates higher than 98%, except for two lung cases with gamma pass rates of 86.9% and 90.6%. The low gamma pass rates showed larger global motion ranges resulting from the baseline shift during beam delivery. Conclusion The actual delivered dose in thoracic and abdominal VMAT under breathing motion was verified by 4D dose reconstruction using typical treatment equipment and software. The proposed method provides a verification method for the actual delivered dose and could be a dosimetric verification QA tool for radiation treatment under various respiratory management techniques.
Collapse
|
64
|
Wolfe AR, Williams TM. Altering the response to radiation: radiosensitizers and targeted therapies in pancreatic ductal adenocarcinoma: preclinical and emerging clinical evidence. ACTA ACUST UNITED AC 2018; 1. [PMID: 32656528 DOI: 10.21037/apc.2018.08.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Radiation therapy continues to have an evolving role in pancreatic ductal adenocarcinoma. While metastatic failure likely contributes to the majority of patient mortality, achieving local control through surgery and/or radiation appears to be important as certain studies suggest that mortality is contributed by local failure. Many studies support that pancreatic cancer is a relatively radiation resistant tumor type. In addition, the ability to further improve radiation through dose escalation strategies in the non-metastatic setting is hampered by closeness of normal organs, including small bowel and stomach, to the tumor. Thus subverting molecular pathways that promote radiation resistance will be critical to further success of radiation in this disease. There is a wealth of preclinical data supporting the targeting of various molecular pathways in combination with radiation therapy, including DNA repair, cell cycle checkpoint proteins, receptor tyrosine kinases, oncoproteins, stem cells, and immunomodulation. A number of clinical trials have been completed or are on-going with novel molecular inhibitors. In this review, we summarize existing preclinical and clinical molecular strategies for improving the efficacy of radiation in pancreatic cancer, and highlight recent and ongoing clinical trials combining radiation and various targeted therapies.
Collapse
Affiliation(s)
- Adam R Wolfe
- Department of Radiation Oncology, The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, OH 43210, USA
| | - Terence M Williams
- Department of Radiation Oncology, The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, OH 43210, USA
| |
Collapse
|
65
|
Teo P, Wang X, Chen B, Zhang H, Yang X, Huang Y, Tang J. Complex of TNF-α and Modified Fe 3O 4 Nanoparticles Suppresses Tumor Growth by Magnetic Induction Hyperthermia. Cancer Biother Radiopharm 2018; 32:379-386. [PMID: 29265918 DOI: 10.1089/cbr.2017.2404] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Magnetic nanoparticles are increasingly applied in clinical area for drug delivery, also in magnetic induction hyperthermia (MIH), and so on. The present research would prepare appropriately modified superparamagnetic iron oxide nanoparticles (SPIONs) to conduct MIH and transfect the anticancer cytokine TNF-α into cells. MATERIALS AND METHODS The SPIONs were surface-modified by 3-aminopropyltriethoxysilane (APTS) and/or protamine sulfate (PRO). The combined MIH using SPIONs and gene therapy were applied to treat Hep G2 cells and tumor model transplanted in nude mice. RESULTS The PRO-SPIONs (the surfaces were sequentially modified by APTS and PRO) showed high transfection efficiency for TNF-α gene with no obvious cytotoxicity. It also exhibited great temperature rising performance under alternating current magnetic field. The combined MIH and gene therapy using PRO-SPIONs/TNF-α complex could reduce cell variability of Hep G2 cells and tumor size transplanted in nude mice. CONCLUSIONS The PRO-SPIONs efficiently transfect the TNF-α gene into Hep G2 cells. Cells expressed more TNF-α when they were exposed to alternating current magnetic field only once. Combined MIH and gene therapy for treatment in vivo exhibited better effects than MIH or gene therapy alone. The combined MIH and gene therapy is promising in liver cancer treatment.
Collapse
Affiliation(s)
- Peishan Teo
- 1 Key Laboratory of Particle & Radiation Imaging of Ministry of Education, Department of Engineering Physics, Tsinghua University , Beijing, People's Republic of China
| | - Xiaowen Wang
- 2 Yuquan Hospital, Medical Center, Tsinghua University , Beijing, People's Republic of China
| | - Benke Chen
- 1 Key Laboratory of Particle & Radiation Imaging of Ministry of Education, Department of Engineering Physics, Tsinghua University , Beijing, People's Republic of China .,3 Department of Biological Pharmaceuticals, Beijing University of Chinese Medicine , Beijing, People's Republic of China
| | - Han Zhang
- 1 Key Laboratory of Particle & Radiation Imaging of Ministry of Education, Department of Engineering Physics, Tsinghua University , Beijing, People's Republic of China .,4 Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Sciences, Shandong University , Jinan, People's Republic of China
| | - Xin Yang
- 1 Key Laboratory of Particle & Radiation Imaging of Ministry of Education, Department of Engineering Physics, Tsinghua University , Beijing, People's Republic of China .,3 Department of Biological Pharmaceuticals, Beijing University of Chinese Medicine , Beijing, People's Republic of China
| | - Yun Huang
- 1 Key Laboratory of Particle & Radiation Imaging of Ministry of Education, Department of Engineering Physics, Tsinghua University , Beijing, People's Republic of China
| | - Jintian Tang
- 1 Key Laboratory of Particle & Radiation Imaging of Ministry of Education, Department of Engineering Physics, Tsinghua University , Beijing, People's Republic of China
| |
Collapse
|
66
|
Nattress CB, Halldén G. Advances in oncolytic adenovirus therapy for pancreatic cancer. Cancer Lett 2018; 434:56-69. [PMID: 29981812 DOI: 10.1016/j.canlet.2018.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 02/06/2023]
Abstract
Survival rates for pancreatic cancer patients have remained unchanged for the last four decades. The most aggressive, and most common, type of pancreatic cancer is pancreatic ductal adenocarcinoma (PDAC), which has the lowest 5-year survival rate of all cancers globally. The poor prognosis is typically due to late presentation of often non-specific symptoms and rapid development of resistance to all current therapeutics, including the standard-of-care cytotoxic drug gemcitabine. While early surgical intervention can significantly prolong patient survival, there are few treatment options for late-stage non-resectable metastatic disease, resulting in mostly palliative care. In addition, a defining feature of pancreatic cancer is the immunosuppressive and impenetrable desmoplastic stroma that blocks access to tumour cells by therapeutic drugs. The limited effectiveness of conventional chemotherapeutics reveals an urgent need to develop novel therapies with different mechanisms of action for this malignancy. An emerging alternative to current therapeutics is oncolytic adenoviruses; these engineered biological agents have proven efficacy and tumour-selectivity in preclinical pancreatic cancer models, including models of drug-resistant cancer. Safety of oncolytic adenoviral mutants has been extensively assessed in clinical trials with only limited toxicity to normal healthy tissue being reported. Promising efficacy in combination with gemcitabine was demonstrated in preclinical and clinical studies. A recent surge in novel adenoviral mutants entering clinical trials for pancreatic cancer indicates improved efficacy through activation of the host anti-tumour responses. The potential for adenoviruses to synergise with chemotherapeutics, activate anti-tumour immune responses, and contribute to stromal dissemination render these mutants highly attractive candidates for improved patient outcomes. Currently, momentum is gathering towards the development of systemically-deliverable mutants that are able to overcome anti-viral host immune responses, erythrocyte binding and hepatic uptake, to promote elimination of primary and metastatic lesions. This review will cover the key components of pancreatic cancer oncogenesis; novel oncolytic adenoviruses; clinical trials; and the current progress in overcoming the challenges of systemic delivery.
Collapse
Affiliation(s)
- Callum Baird Nattress
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, United Kingdom
| | - Gunnel Halldén
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, United Kingdom.
| |
Collapse
|
67
|
Moradi Marjaneh R, Hassanian SM, Ghobadi N, Ferns GA, Karimi A, Jazayeri MH, Nasiri M, Avan A, Khazaei M. Targeting the death receptor signaling pathway as a potential therapeutic target in the treatment of colorectal cancer. J Cell Physiol 2018; 233:6538-6549. [DOI: 10.1002/jcp.26640] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 03/30/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Reyhaneh Moradi Marjaneh
- Department of Physiology, Faculty 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, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Microanatomy Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Niloofar Ghobadi
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Gordon A. Ferns
- Brighton & Sussex Medical School Division of Medical Education Falmer, Brighton, Sussex UK
| | - Afshin Karimi
- Quality Department of Nutricia Mashhad Mild Powder Industrial Mashhad Iran
| | - Mir Hadi Jazayeri
- Immunology Research Center and Department of Immunology, School of Medicine Iran University of Medical Sciences Tehran Iran
| | - Mohammadreza Nasiri
- Recombinant Proteins Research Group The Research Institute of Biotechnology, Ferdowsi University of Mashhad Mashhad Iran
| | - Amir Avan
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
- Cancer Research Center Mashhad University of Medical Sciences Mashhad Iran
- Department of Modern Sciences and Technologies, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Surgical Oncology Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
68
|
Therapeutic activity of retroviral replicating vector-mediated prodrug activator gene therapy for pancreatic cancer. Cancer Gene Ther 2018; 25:184-195. [PMID: 29735994 DOI: 10.1038/s41417-018-0020-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/17/2018] [Indexed: 01/20/2023]
Abstract
Toca 511, a retroviral replicating vector (RRV) encoding the yeast cytosine deaminase (yCD) prodrug activator gene, which mediates conversion of the prodrug 5-fluorocytosine (5-FC) to the anticancer drug 5-fluorouracil (5-FU), is currently being evaluated in Phase II/III clinical trials for glioma, and showing highly promising evidence of therapeutic activity. Here we evaluated RRV-mediated prodrug activator gene therapy as a new therapeutic approach for pancreatic ductal adenocarcinoma (PDAC). RRV spread rapidly and conferred significant cytotoxicity with prodrug in a panel of PDAC cells. Efficient intratumoral replication and complete inhibition of tumor growth upon 5-FC administration were observed in both immunodeficient and immunocompetent subcutaneous PDAC models. Biodistribution of RRV was highly restricted in normal tissues, especially in immunocompetent hosts. Tumor growth inhibition by Toca 511 followed by 5-FC was also confirmed in the orthotopic PDAC model. This study provides the first proof-of-concept for application of Toca 511 and Toca FC (extended release 5-FC) to the treatment of human PDAC, and provided support for inclusion of PDAC in a Phase I study evaluating Toca 511 in various systemic malignancies, (NCT02576665), which has recently been initiated.
Collapse
|
69
|
Han H, Wang L, Xu J, Wang A. miR-128 induces pancreas cancer cell apoptosis by targeting MDM4. Exp Ther Med 2018; 15:5017-5022. [PMID: 29805525 DOI: 10.3892/etm.2018.6047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 02/08/2018] [Indexed: 01/17/2023] Open
Abstract
MicroRNAs (miRNA/miRs) are small, non-coding RNA molecules (19-25 nucleotides in length), which function to regulate gene expression. It has been reported that miR-128 serves an important role in regulating cancer cell growth; increasing evidence has indicated that the expression of miR-128 is decreased in pancreatic cancer (PC) cells. However, the specific mechanisms of miR-128 in regulating PC cell growth are unclear. In the present study, it was confirmed that the expression of miR-128 was significantly decreased within PC tissues compared with adjacent normal tissues via reverse transcription-quantitative polymerase chain reaction analysis. In addition, miR-128 mimics inhibited PC MIA-PaCa2 cell growth by enhancing cell apoptosis in a caspase-dependent manner. Furthermore, the results of the present study demonstrated that double minute 4 (MDM4) may be a direct target for miR-128 via a dual luciferase report assay; miR-128 may inhibit MDM4 expression, and increase p53 and cleaved caspase-3 protein expression levels. In summary, the present study indicated that miR-128 is downregulated in PC, and it may be a promising target for future PC diagnosis and treatment.
Collapse
Affiliation(s)
- Hongchao Han
- Department of General Surgery, The Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Lisheng Wang
- Department of General Surgery, The Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Jie Xu
- Department of Gynecology and Obstetrics, The Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Aikun Wang
- Department of General Surgery, The Third People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| |
Collapse
|
70
|
Chin V, Nagrial A, Sjoquist K, O'Connor CA, Chantrill L, Biankin AV, Scholten RJPM, Yip D, Cochrane Upper GI and Pancreatic Diseases Group. Chemotherapy and radiotherapy for advanced pancreatic cancer. Cochrane Database Syst Rev 2018; 3:CD011044. [PMID: 29557103 PMCID: PMC6494171 DOI: 10.1002/14651858.cd011044.pub2] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic cancer (PC) is a highly lethal disease with few effective treatment options. Over the past few decades, many anti-cancer therapies have been tested in the locally advanced and metastatic setting, with mixed results. This review attempts to synthesise all the randomised data available to help better inform patient and clinician decision-making when dealing with this difficult disease. OBJECTIVES To assess the effect of chemotherapy, radiotherapy or both for first-line treatment of advanced pancreatic cancer. Our primary outcome was overall survival, while secondary outcomes include progression-free survival, grade 3/4 adverse events, therapy response and quality of life. SEARCH METHODS We searched for published and unpublished studies in CENTRAL (searched 14 June 2017), Embase (1980 to 14 June 2017), MEDLINE (1946 to 14 June 2017) and CANCERLIT (1999 to 2002) databases. We also handsearched all relevant conference abstracts published up until 14 June 2017. SELECTION CRITERIA All randomised studies assessing overall survival outcomes in patients with advanced pancreatic ductal adenocarcinoma. Chemotherapy and radiotherapy, alone or in combination, were the eligible treatments. DATA COLLECTION AND ANALYSIS Two review authors independently analysed studies, and a third settled any disputes. We extracted data on overall survival (OS), progression-free survival (PFS), response rates, adverse events (AEs) and quality of life (QoL), and we assessed risk of bias for each study. MAIN RESULTS We included 42 studies addressing chemotherapy in 9463 patients with advanced pancreatic cancer. We did not identify any eligible studies on radiotherapy.We did not find any benefit for chemotherapy over best supportive care. However, two identified studies did not have sufficient data to be included in the analysis, and many of the chemotherapy regimens studied were outdated.Compared to gemcitabine alone, participants receiving 5FU had worse OS (HR 1.69, 95% CI 1.26 to 2.27, moderate-quality evidence), PFS (HR 1.47, 95% CI 1.12 to 1.92) and QoL. On the other hand, two studies showed FOLFIRINOX was better than gemcitabine for OS (HR 0.51 95% CI 0.43 to 0.60, moderate-quality evidence), PFS (HR 0.46, 95% CI 0.38 to 0.57) and response rates (RR 3.38, 95% CI 2.01 to 5.65), but it increased the rate of side effects. The studies evaluating CO-101, ZD9331 and exatecan did not show benefit or harm when compared with gemcitabine alone.Giving gemcitabine at a fixed dose rate improved OS (HR 0.79, 95% CI 0.66 to 0.94, high-quality evidence) but increased the rate of side effects when compared with bolus dosing.When comparing gemcitabine combinations to gemcitabine alone, gemcitabine plus platinum improved PFS (HR 0.80, 95% CI 0.68 to 0.95) and response rates (RR 1.48, 95% CI 1.11 to 1.98) but not OS (HR 0.94, 95% CI 0.81 to 1.08, low-quality evidence). The rate of side effects increased. Gemcitabine plus fluoropyrimidine improved OS (HR 0.88, 95% CI 0.81 to 0.95), PFS (HR 0.79, 95% CI 0.72 to 0.87) and response rates (RR 1.78, 95% CI 1.29 to 2.47, high-quality evidence), but it also increased side effects. Gemcitabine plus topoisomerase inhibitor did not improve survival outcomes but did increase toxicity. One study demonstrated that gemcitabine plus nab-paclitaxel improved OS (HR 0.72, 95% CI 0.62 to 0.84, high-quality evidence), PFS (HR 0.69, 95% CI 0.58 to 0.82) and response rates (RR 3.29, 95% CI 2.24 to 4.84) but increased side effects. Gemcitabine-containing multi-drug combinations (GEMOXEL or cisplatin/epirubicin/5FU/gemcitabine) improved OS (HR 0.55, 95% CI 0.39 to 0.79, low-quality evidence), PFS (HR 0.43, 95% CI 0.30 to 0.62) and QOL.We did not find any survival advantages when comparing 5FU combinations to 5FU alone. AUTHORS' CONCLUSIONS Combination chemotherapy has recently overtaken the long-standing gemcitabine as the standard of care. FOLFIRINOX and gemcitabine plus nab-paclitaxel are highly efficacious, but our analysis shows that other combination regimens also offer a benefit. Selection of the most appropriate chemotherapy for individual patients still remains difficult, with clinicopathological stratification remaining elusive. Biomarker development is essential to help rationalise treatment selection for patients.
Collapse
Affiliation(s)
- Venessa Chin
- Garvan Institute of Medical ResearchThe Kinghorn Cancer Centre384 Victoria Street DarlinghurstSydneyNSWAustralia2010
- St Vincent's HospitalSydneyNSWAustralia
| | - Adnan Nagrial
- Garvan Institute of Medical ResearchThe Kinghorn Cancer Centre384 Victoria Street DarlinghurstSydneyNSWAustralia2010
- The Crown Princess Mary Cancer CentreDarcy RoadWestmeadNSWAustralia2145
| | - Katrin Sjoquist
- University of SydneyNHMRC Clinical Trials CentreK25 ‐ Medical Foundation BuildingSydneyNSWAustralia2006
- Cancer Care Centre, St George HospitalMedical OncologySt George Hospital, Gray StKogarahAustraliaNSW 2217
| | - Chelsie A O'Connor
- St Vincent's HospitalSydneyNSWAustralia
- Genesis Cancer CareSydneyNSWAustralia
- Macquarie University HospitalSydneyAustralia
| | - Lorraine Chantrill
- The Kinghorn Cancer Centre, Garvan Institute of Medical ResearchDepartment of Pancreatic Cancer382 Victoria Street DarlinghurstSydneyNSWAustralia2010
| | - Andrew V Biankin
- University of GlasgowInstitute of Cancer SciencesWolfson Wohl Cancer Research CentreGarscube Estate, Switchback RoadGlasgowUKG61 1QH
- University of New South WalesSouth Western Sydney Clinical School, Faculty of MedicineLiverpoolNSWAustralia2170
- West of Scotland Pancreatic Unit and Glasgow Royal InfirmaryGlasgowUK
| | - Rob JPM Scholten
- Julius Center for Health Sciences and Primary Care / University Medical Center UtrechtCochrane NetherlandsRoom Str. 6.126P.O. Box 85500UtrechtNetherlands3508 GA
| | - Desmond Yip
- The Canberra HospitalDepartment of Medical OncologyYamba DriveGarranACTAustralia2605
- Australian National UniversityANU Medical SchoolActonACTAustralia0200
| | | |
Collapse
|
71
|
Yang L, Yang G, Ding Y, Dai Y, Xu S, Guo Q, Xie A, Hu G. Inhibition of PI3K/AKT Signaling Pathway Radiosensitizes Pancreatic Cancer Cells with ARID1A Deficiency in Vitro. J Cancer 2018; 9:890-900. [PMID: 29581767 PMCID: PMC5868153 DOI: 10.7150/jca.21306] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 01/29/2018] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer is among the most aggressive human cancers, and is resistant to regular chemotherapy and radiotherapy. The AT-rich interactive domain containing protein 1A (ARID1A) gene, a crucial chromatin remodeling gene, mutates frequently in a broad spectrum of cancers, including pancreatic cancer. Recent evidence suggests that ARID1A acts as tumor suppressor and plays an important role in DNA damage repair (DDR). However, the effect of ARID1A on the radiosensitivity of pancreatic cancer remains unclear. Herein, we investigated the involvement of ARID1A depletion in the radioresistance of pancreatic cancer cells, and explored the underlying mechanisms. The results reveal that knockdown of ARID1A enhances the radioresistance of pancreatic cancer cells through suppressing apoptosis, impairing G2-M checkpoint arrest, strengthening DDR, and accompanying activation of PI3K/AKT signaling pathway. Moreover, upon inhibition of PI3K/AKT pathway by PI3K-inhibitor LY294002 or AKT-inhibitor mk2206, the radiosensitivity of ARID1A-deficient pancreatic cancer cells is improved in vitro via increased apoptosis and weakened DDR. Taken together, these data suggest that loss of ARID1A expression enhances radioresistance of pancreatic cancer through activation of PI3K/AKT pathway, which maybe a promising target for radiosensitization of ARID1A-deficient pancreatic cancer.
Collapse
Affiliation(s)
- Lin Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Guanghai Yang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yingjun Ding
- Department of Physiology, University of Oklahoma Health Sciences Center, 975 N 10 th St, Oklahoma City, OK, US, 73104
| | - Yuhong Dai
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Sanpeng Xu
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qiuyun Guo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Aini Xie
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
72
|
Kamensek U, Cemazar M, Lampreht Tratar U, Ursic K, Sersa G. Antitumor in situ vaccination effect of TNFα and IL-12 plasmid DNA electrotransfer in a murine melanoma model. Cancer Immunol Immunother 2018; 67:785-795. [PMID: 29468364 PMCID: PMC5928174 DOI: 10.1007/s00262-018-2133-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 02/13/2018] [Indexed: 12/15/2022]
Abstract
Gene electrotransfer (GET) is one of the most efficient non-viral gene therapy approaches for the localized transfer of multiple genes into tumors in vivo; therefore, it is especially promising for delivering different cytokines that are toxic if administered systemically. In this study, we used concomitant intratumoral GET of two cytokines: tumor necrosis factor alpha (TNFα), a potent cytotoxic cytokine to induce in situ vaccination, and interleukin 12 (IL-12), an immunostimulatory cytokine to boost the primed local immune response into a systemic one. After performing GET in murine melanoma tumors, both TNFα and IL-12 mRNA levels were significantly increased, which resulted in a pronounced delay in tumor growth of 27 days and a prolonged survival time of mice. An antitumor immune response was confirmed by extensive infiltration of immune cells in the tumor site, and expansion of the effector immune cells in the sentinel lymph nodes. Furthermore, the effect of in situ vaccination was indicated by the presence of vitiligo localized to the treatment area and resistance of the mice to secondary challenge with tumor cells. Intratumoral GET of two cytokines, one for in situ vaccination and one for an immune boost, proved feasible and effective in eliciting a potent and durable antitumor response; therefore, further studies of this approach are warranted.
Collapse
Affiliation(s)
- Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, 1000, Ljubljana, Slovenia.
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, 1000, Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, Polje 42, Izola, Slovenia
| | - Ursa Lampreht Tratar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, 1000, Ljubljana, Slovenia
| | - Katja Ursic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, 1000, Ljubljana, Slovenia
| | - Gregor Sersa
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, 1000, Ljubljana, Slovenia
- Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, 1000, Ljubljana, Slovenia
| |
Collapse
|
73
|
Ogawa K, Kamimura K, Kobayashi Y, Abe H, Yokoo T, Sakai N, Nagoya T, Sakamaki A, Abe S, Hayashi K, Ikarashi S, Kohisa J, Tsuchida M, Aoyagi Y, Zhang G, Liu D, Terai S. Efficacy and Safety of Pancreas-Targeted Hydrodynamic Gene Delivery in Rats. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 9:80-88. [PMID: 29246326 PMCID: PMC5612811 DOI: 10.1016/j.omtn.2017.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/14/2017] [Accepted: 08/14/2017] [Indexed: 12/18/2022]
Abstract
Development of an effective, safe, and convenient method for gene delivery to the pancreas is a critical step toward gene therapy for pancreatic diseases. Therefore, we tested the possibility of applying the principle of hydrodynamic gene delivery for successful gene transfer to pancreas using rats as a model. The established procedure involves the insertion of a catheter into the superior mesenteric vein with temporary blood flow occlusion at the portal vein and hydrodynamic injection of DNA solution. We demonstrated that our procedure achieved efficient pancreas-specific gene expression that was 2,000-fold higher than that seen in the pancreas after the systemic hydrodynamic gene delivery. In addition, the level of gene expression achieved in the pancreas by the pancreas-specific gene delivery was comparable to the level in the liver achieved by a liver-specific hydrodynamic gene delivery. The optimal level of reporter gene expression in the pancreas requires an injection volume equivalent to 2.0% body weight with flow rate of 1 mL/s and plasmid DNA concentration at 5 μg/mL. With the exception of transient expansion of intercellular spaces and elevation of serum amylase levels, which recovered within 3 days, no permanent tissue damage was observed. These results suggest that pancreas-targeted hydrodynamic gene delivery is an effective and safe method for gene delivery to the pancreas and clinically applicable.
Collapse
Affiliation(s)
- Kohei Ogawa
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
| | - Yuji Kobayashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiroyuki Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takeshi Yokoo
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Norihiro Sakai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takuro Nagoya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Akira Sakamaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Satoshi Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kazunao Hayashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Satoshi Ikarashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Junji Kohisa
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masanori Tsuchida
- Division of Thoracic and Cardiovascular Surgery, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yutaka Aoyagi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Guisheng Zhang
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Dexi Liu
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
74
|
Lee LS, Andersen DK, Ashida R, Brugge WR, Canto MI, Chang KJ, Chari ST, DeWitt J, Hwang JH, Khashab MA, Kim K, Levy MJ, McGrath K, Park WG, Singhi A, Stevens T, Thompson CC, Topazian MD, Wallace MB, Wani S, Waxman I, Yadav D, Singh VK. EUS and related technologies for the diagnosis and treatment of pancreatic disease: research gaps and opportunities-Summary of a National Institute of Diabetes and Digestive and Kidney Diseases workshop. Gastrointest Endosc 2017; 86:768-778. [PMID: 28941651 PMCID: PMC6698378 DOI: 10.1016/j.gie.2017.08.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 12/11/2022]
Abstract
A workshop was sponsored by the National Institute of Diabetes and Digestive and Kidney Diseases to address the research gaps and opportunities in pancreatic EUS. The event occurred on July 26, 2017 in 4 sessions: (1) benign pancreatic diseases, (2) high-risk pancreatic diseases, (3) diagnostic and therapeutics, and (4) new technologies. The current state of knowledge was reviewed, with identification of numerous gaps in knowledge and research needs. Common themes included the need for large multicenter consortia of various pancreatic diseases to facilitate meaningful research of these entities; to standardize EUS features of different pancreatic disorders, the technique of sampling pancreatic lesions, and the performance of various therapeutic EUS procedures; and to identify high-risk disease early at the cellular level before macroscopic disease develops. The need for specialized tools and accessories to enable the safe and effective performance of therapeutic EUS procedures also was discussed.
Collapse
Affiliation(s)
- Linda S Lee
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Dana K Andersen
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Reiko Ashida
- Departments of Cancer Survey and Gastrointestinal Oncology, Osaka Prefectural Hospital Organization, Osaka International Cancer Institute, Osaka, Japan
| | - William R Brugge
- Department of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mimi I Canto
- Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kenneth J Chang
- Comprehensive Digestive Disease Center, Department of Gastroenterology and Hepatology, University of California at Irvine Health, Orange, California, USA
| | - Suresh T Chari
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - John DeWitt
- Division of Gastroenterology, Indiana University Health Medical Center, Indianapolis, Indiana, USA
| | - Joo Ha Hwang
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Mouen A Khashab
- Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kang Kim
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael J Levy
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kevin McGrath
- Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Walter G Park
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Aatur Singhi
- Department of Pathology, University of Pittsburgh Medical Center, Sewickley, Pennsylvania, USA
| | - Tyler Stevens
- Department of Gastroenterology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Christopher C Thompson
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mark D Topazian
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael B Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Sachin Wani
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Irving Waxman
- Department of Medicine, The University of Chicago Comprehensive Cancer Center, University of Chicago School of Medicine, Chicago, Illinois, USA
| | - Dhiraj Yadav
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Vikesh K Singh
- Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
75
|
Koklu S, Basar O, Brugge WR. Endoscopic ultrasound guided tumor directed therapy and cyst ablation. TECHNIQUES IN GASTROINTESTINAL ENDOSCOPY 2017. [DOI: 10.1016/j.tgie.2017.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
76
|
Baird JR, Monjazeb AM, Shah O, McGee H, Murphy WJ, Crittenden MR, Gough MJ. Stimulating Innate Immunity to Enhance Radiation Therapy-Induced Tumor Control. Int J Radiat Oncol Biol Phys 2017; 99:362-373. [PMID: 28871985 PMCID: PMC5604475 DOI: 10.1016/j.ijrobp.2017.04.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/02/2017] [Indexed: 12/29/2022]
Abstract
Novel ligands that target Toll-like receptors and other innate recognition pathways represent a potent strategy for modulating innate immunity to generate antitumor immunity. Although many of the current clinically successful immunotherapies target adaptive T-cell responses, both preclinical and clinical studies suggest that adjuvants have the potential to enhance the scope and efficacy of cancer immunotherapy. Radiation may be a particularly good partner to combine with innate immune therapies, because it is a highly efficient means to kill cancer cells but may fail to send the appropriate inflammatory signals needed to act as an efficient endogenous vaccine. This may explain why although radiation therapy is a highly used cancer treatment, true abscopal effects-regression of disease outside the field without additional systemic therapy-are extremely rare. This review focuses on efforts to combine innate immune stimuli as adjuvants with radiation, creating a distinct and complementary approach from T cell-targeted therapies to enhance antitumor immunity.
Collapse
Affiliation(s)
- Jason R Baird
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, Oregon
| | - Arta M Monjazeb
- Department of Radiation Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California; Laboratory of Cancer Immunology, UC Davis Comprehensive Cancer Center, Sacramento, California
| | - Omid Shah
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Heather McGee
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - William J Murphy
- Laboratory of Cancer Immunology, UC Davis Comprehensive Cancer Center, Sacramento, California
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, Oregon; The Oregon Clinic, Portland, Oregon
| | - Michael J Gough
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, Oregon.
| |
Collapse
|
77
|
Mukewar S, Muthusamy VR. Recent Advances in Therapeutic Endosonography for Cancer Treatment. Gastrointest Endosc Clin N Am 2017; 27:657-680. [PMID: 28918804 DOI: 10.1016/j.giec.2017.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Therapeutic endosonography (EUS) may play an important role in the management of cancers. EUS-guided fiducial placement has a high success rate and can aid in stereotactic radiotherapy. EUS-guided tumor ablation therapies can help in palliation of locally advanced tumors. EUS-guided antitumor injection seems to be feasible and safe in animals; initial human studies suffer from small sample size and lack of controls. Randomized, controlled trials have not shown benefit over conventional therapy. EUS celiac plexus neurolysis has gained popularity and is performed by interventional endosonographers. Large trials are needed to determine the most appropriate indications and overall usefulness of these therapies.
Collapse
Affiliation(s)
- Saurabh Mukewar
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, 200 UCLA Medical Plaza, Room 330-37, Los Angeles, CA 90095, USA
| | - Venkataraman Raman Muthusamy
- The Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, 200 UCLA Medical Plaza, Room 330-37, Los Angeles, CA 90095, USA.
| |
Collapse
|
78
|
Hua YQ, Wang P, Zhu XY, Shen YH, Wang K, Shi WD, Lin JH, Meng ZQ, Chen Z, Chen H. Radiofrequency ablation for hepatic oligometastatic pancreatic cancer: An analysis of safety and efficacy. Pancreatology 2017; 17:967-973. [PMID: 29129384 DOI: 10.1016/j.pan.2017.08.072] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 08/17/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVES This study was to evaluate the value of radiofrequency ablation (RFA) in the treatment of pancreatic cancer with synchronous liver oligometastasis. METHODS 102 patients diagnosed with pancreatic cancer with synchronous liver oligometastasis undergoing RFA were recruited in this retrospective study between January 2012 and December 2015. Clinical efficacy was evaluated by computed tomography or magnetic resonance imaging 1 month later. All patients were treated with RFA and systemic chemotherapy based on NCCN guideline. RESULTS The median follow-up was 21 months (range, 4.0-43.8 months). Of all patients, the 1-year survival rate was 47.1% and the median overall survival time was 11.40 months. Complete tumor ablation was achieved in 137 of 145 RFA sessions (94.5%), and in 244 of 254 tumors (96.1%). The incidence of common complications was 9.8%, and no severe complications were reported in any patient. Multivariate Cox regression analysis revealed that primary tumor in the head of the pancreas (HR = 1.868, 95% CI: 1.023-3.409; P = 0.042), maximum diameter of liver metastasis 3-5 cm (HR = 1.801, 95% CI: 1.081-3.001, P = 0.024) and neutrophil/lymphocyte ratio (NLR) ≥2.5 (HR = 1.716, 95% CI: 1.047-2.811; P = 0.032) were independent predictors of poorer survival. CONCLUSION RFA provides a minimally invasive and safe treatment for patients with pancreatic cancer with liver oligometastases. The clinical efficiency of RFA for hepatic oligometastatic pancreatic cancer was easily affected by the following factors: primary tumor location, maximum diameter of liver metastasis and NLR. These factors could be helpful for treatment decision and clinical trial design.
Collapse
Affiliation(s)
- Yong-Qiang Hua
- Minimally Invasive Treatment Center, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong An Road, Shanghai 200032, PR China
| | - Peng Wang
- Minimally Invasive Treatment Center, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai 200032, PR China
| | - Xiao-Yan Zhu
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong An Road, Shanghai 200032, PR China
| | - Ye-Hua Shen
- Minimally Invasive Treatment Center, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai 200032, PR China
| | - Kun Wang
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong An Road, Shanghai 200032, PR China
| | - Wei-Dong Shi
- Minimally Invasive Treatment Center, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai 200032, PR China
| | - Jun-Hua Lin
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong An Road, Shanghai 200032, PR China
| | - Zhi-Qiang Meng
- Minimally Invasive Treatment Center, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai 200032, PR China
| | - Zhen Chen
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong An Road, Shanghai 200032, PR China
| | - Hao Chen
- Minimally Invasive Treatment Center, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong An Road, Shanghai 200032, PR China.
| |
Collapse
|
79
|
Rao AD, Feng Z, Shin EJ, He J, Waters KM, Coquia S, DeJong R, Rosati LM, Su L, Li D, Jackson J, Clark S, Schultz J, Hutchings D, Kim SH, Hruban RH, DeWeese TL, Wong J, Narang A, Herman JM, Ding K. A Novel Absorbable Radiopaque Hydrogel Spacer to Separate the Head of the Pancreas and Duodenum in Radiation Therapy for Pancreatic Cancer. Int J Radiat Oncol Biol Phys 2017; 99:1111-1120. [PMID: 28943075 DOI: 10.1016/j.ijrobp.2017.08.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 01/25/2023]
Abstract
PURPOSE We assessed the feasibility and theoretical dosimetric advantages of an injectable hydrogel to increase the space between the head of the pancreas (HOP) and duodenum in a human cadaveric model. METHODS AND MATERIALS Using 3 human cadaveric specimens, an absorbable radiopaque hydrogel was injected between the HOP and duodenum by way of open laparotomy in 1 case and endoscopic ultrasound (EUS) guidance in 2 cases. The cadavers were subsequently imaged using computed tomography and dissected for histologic confirmation of hydrogel placement. The duodenal dose reduction and planning target volume (PTV) coverage were characterized using pre- and postspacer injection stereotactic body radiation therapy (SBRT) plans for the 2 cadavers with EUS-guided placement, the delivery method that appeared the most clinically desirable. Modeling studies were performed using 60 SBRT plans consisting of 10 previously treated patients with unresectable pancreatic cancer, each with 6 different HOP-duodenum separation distances. The duodenal volume receiving 15 Gy (V15), 20 Gy (V20), and 33 Gy (V33) was assessed for each iteration. RESULTS In the 3 cadaveric studies, an average of 0.9 cm, 1.1 cm, and 0.9 cm HOP-duodenum separation was achieved. In the 2 EUS cases, the V20 decreased from 3.86 cm3 to 0.36 cm3 and 3.75 cm3 to 1.08 cm3 (treatment constraint <3 cm3), and the V15 decreased from 7.07 cm3 to 2.02 cm3 and 9.12 cm3 to 3.91 cm3 (treatment constraint <9 cm3). The PTV coverage improved or was comparable between the pre- and postinjection studies. Modeling studies demonstrated that a separation of 8 mm was sufficient to consistently reduce the V15, V20, and V33 to acceptable clinical constraints. CONCLUSIONS Currently, dose escalation has been limited owing to radiosensitive structures adjacent to the pancreas. We demonstrated the feasibility of hydrogel separation of the HOP and duodenum. Future studies will evaluate the safety and efficacy of this technique with the potential for more effective dose escalation using SBRT or intensity-modulated radiation therapy to improve the outcomes in patients with unresectable pancreatic cancer.
Collapse
Affiliation(s)
- Avani D Rao
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ziwei Feng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Eun Ji Shin
- Department of Gastroenterology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jin He
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Kevin M Waters
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Stephanie Coquia
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Robert DeJong
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Lauren M Rosati
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Lin Su
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Dengwang Li
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Juan Jackson
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Stephen Clark
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jeffrey Schultz
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Danielle Hutchings
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Seong-Hun Kim
- Department of Gastroenterology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ralph H Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Theodore L DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - John Wong
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Amol Narang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Joseph M Herman
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Kai Ding
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
80
|
The Emerging Role of Endoscopists in Treating Unresectable Pancreatic Cancer. Pancreas 2017; 46:839-849. [PMID: 28697122 DOI: 10.1097/mpa.0000000000000862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Pancreatic adenocarcinoma is the eighth leading cause of cancer deaths worldwide in men and ninth leading cause in women. Surgical resection offers the only chance of potential cure; however, only 9.4% of patients present at the localized, resectable stage, whereas the rest present at the locally advanced or metastatic, unresectable stages. Because of the guarded outcomes following systemic chemoradiation and the associated systemic toxicities, locoregional therapies have recently gained popularity. Various endoscopic techniques (endoscopic ultrasound [EUS]-guided ablative therapies, fine-needle instillation of antitumor agents, stereotactic body radiation therapy with EUS-guided fiducial marker placement, and EUS-guided brachytherapy) have been explored over the past several years. Endoscopic therapy plays a role in the treatment of unresectable pancreatic adenocarcinoma. Its minimal invasiveness and increased precision of delivering oncologic treatments under EUS guidance render it as a favorable option for patients who do not benefit from surgical resection. New endoscopic therapies are currently under investigation, and the emerging role of the endoscopist in the treatment of unresectable pancreatic cancer continues to grow.
Collapse
|
81
|
EUS-guided paclitaxel injection as an adjunctive therapy to systemic chemotherapy and concurrent external beam radiation before surgery for localized or locoregional esophageal cancer: a multicenter prospective randomized trial. Gastrointest Endosc 2017; 86:140-149. [PMID: 27890801 DOI: 10.1016/j.gie.2016.11.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/03/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS OncoGel (Protherics Salt Lake City, Inc, Salt Lake City, UT) is paclitaxel (PTX) formulated in a thermosensitive, biodegradable copolymer for focused cytotoxicity and radiosensitization. A phase 2a study suggested that EUS-guided PTX injection into esophageal tumors subsequently receiving radiotherapy was safe. METHODS In an international multicenter, prospective, randomized phase 2b study, patients with local or locoregional adenocarcinoma or squamous cell carcinoma (SCC) of the esophagus/gastroesophageal junction and eligible for neoadjuvant chemoradiotherapy (CRT) before surgery were randomized to standard of care (SOC) plus EUS-guided PTX injection or SOC alone. PTX was injected in 0.5 to 1.0 mL aliquots throughout the tumor. Planned CRT as SOC was intravenous 5-fluorouracil for the first 4 days (weeks 1 and 5), intravenous cisplatin on the first day of each 5-fluorouracil course, and radiotherapy over 5.5 weeks. Patients were evaluated weekly during CRT and re-evaluated at 12 weeks for surgical eligibility and CT for change in overall tumor volume. RESULTS The analysis included 137 patients (97 males; mean age, 58 ± 9.1 years) randomized to PTX + SOC (n = 72) and SOC (n = 65) by using a modified intention-to-treat approach. Overall response by tumor volume between the PTX (12.5%) and the SOC group (20.0%; P = .24; odds ratio, 0.57; 95% confidence interval, 0.23-1.44) was similar. Pathologic complete response was higher in the SOC group (26.2% vs 12.5%; P = .046); however, 12-month survival (P = .412) and the overall frequency of 1 or more adverse events (P = .17) were similar between the 2 groups. CONCLUSIONS SOC + PTX is safe but does not improve overall survival or overall tumor response at the primary tumor site for patients with local or locoregional cancer of the esophagus/gastroesophageal junction. (Clinical trial registration number: NCT00573131.).
Collapse
|
82
|
Murphy C, Rettedal E, Lehouritis P, Devoy C, Tangney M. Intratumoural production of TNFα by bacteria mediates cancer therapy. PLoS One 2017; 12:e0180034. [PMID: 28662099 PMCID: PMC5491124 DOI: 10.1371/journal.pone.0180034] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/15/2017] [Indexed: 12/24/2022] Open
Abstract
Systemic administration of the highly potent anticancer therapeutic, tumour necrosis factor alpha (TNFα) induces high levels of toxicity and is responsible for serious side effects. Consequently, tumour targeting is required in order to confine this toxicity within the locality of the tumour. Bacteria have a natural capacity to grow within tumours and deliver therapeutic molecules in a controlled fashion. The non-pathogenic E. coli strain MG1655 was investigated as a tumour targeting system in order to produce TNFα specifically within murine tumours. In vivo bioluminescence imaging studies and ex vivo immunofluorescence analysis demonstrated rapid targeting dynamics and prolonged survival, replication and spread of this bacterial platform within tumours. An engineered TNFα producing construct deployed in mouse models via either intra-tumoural (i.t.) or intravenous (i.v.) administration facilitated robust TNFα production, as evidenced by ELISA of tumour extracts. Tumour growth was impeded in three subcutaneous murine tumour models (CT26 colon, RENCA renal, and TRAMP prostate) as evidenced by tumour volume and survival analyses. A pattern of pro-inflammatory cytokine induction was observed in tumours of treated mice vs. controls. Mice remained healthy throughout experiments. This study indicates the therapeutic efficacy and safety of TNFα expressing bacteria in vivo, highlighting the potential of non-pathogenic bacteria as a platform for restricting the activity of highly potent cancer agents to tumours.
Collapse
Affiliation(s)
- Carola Murphy
- Cork Cancer Research Centre, University College Cork, Cork, Ireland
| | | | - Panos Lehouritis
- Cork Cancer Research Centre, University College Cork, Cork, Ireland
| | - Ciarán Devoy
- Cork Cancer Research Centre, University College Cork, Cork, Ireland
- SynBioCentre, University College Cork, Cork, Ireland
| | - Mark Tangney
- Cork Cancer Research Centre, University College Cork, Cork, Ireland
- SynBioCentre, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
- * E-mail:
| |
Collapse
|
83
|
Rouanet M, Lebrin M, Gross F, Bournet B, Cordelier P, Buscail L. Gene Therapy for Pancreatic Cancer: Specificity, Issues and Hopes. Int J Mol Sci 2017; 18:ijms18061231. [PMID: 28594388 PMCID: PMC5486054 DOI: 10.3390/ijms18061231] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/01/2017] [Accepted: 06/01/2017] [Indexed: 12/13/2022] Open
Abstract
A recent death projection has placed pancreatic ductal adenocarcinoma as the second cause of death by cancer in 2030. The prognosis for pancreatic cancer is very poor and there is a great need for new treatments that can change this poor outcome. Developments of therapeutic innovations in combination with conventional chemotherapy are needed urgently. Among innovative treatments the gene therapy offers a promising avenue. The present review gives an overview of the general strategy of gene therapy as well as the limitations and stakes of the different experimental in vivo models, expression vectors (synthetic and viral), molecular tools (interference RNA, genome editing) and therapeutic genes (tumor suppressor genes, antiangiogenic and pro-apoptotic genes, suicide genes). The latest developments in pancreatic carcinoma gene therapy are described including gene-based tumor cell sensitization to chemotherapy, vaccination and adoptive immunotherapy (chimeric antigen receptor T-cells strategy). Nowadays, there is a specific development of oncolytic virus therapies including oncolytic adenoviruses, herpes virus, parvovirus or reovirus. A summary of all published and on-going phase-1 trials is given. Most of them associate gene therapy and chemotherapy or radiochemotherapy. The first results are encouraging for most of the trials but remain to be confirmed in phase 2 trials.
Collapse
Affiliation(s)
- Marie Rouanet
- Department of Gastroenterology, CHU Rangueil, 1 avenue Jean Poulhès, Toulouse 31059, France.
- INSERM UMR 1037, Cancer Research Center of Toulouse, Toulouse 31037, France.
| | - Marine Lebrin
- Center for Clinical Investigation 1436, Module of Biotherapy, CHU Rangueil, 1 avenue Jean Poulhès, Toulouse Cedex 9, France.
| | - Fabian Gross
- Center for Clinical Investigation 1436, Module of Biotherapy, CHU Rangueil, 1 avenue Jean Poulhès, Toulouse Cedex 9, France.
| | - Barbara Bournet
- Department of Gastroenterology, CHU Rangueil, 1 avenue Jean Poulhès, Toulouse 31059, France.
- INSERM UMR 1037, Cancer Research Center of Toulouse, Toulouse 31037, France.
- University of Toulouse III, Medical School of Medicine Rangueil, Toulouse 31062, France.
| | - Pierre Cordelier
- INSERM UMR 1037, Cancer Research Center of Toulouse, Toulouse 31037, France.
| | - Louis Buscail
- Department of Gastroenterology, CHU Rangueil, 1 avenue Jean Poulhès, Toulouse 31059, France.
- INSERM UMR 1037, Cancer Research Center of Toulouse, Toulouse 31037, France.
- Center for Clinical Investigation 1436, Module of Biotherapy, CHU Rangueil, 1 avenue Jean Poulhès, Toulouse Cedex 9, France.
- University of Toulouse III, Medical School of Medicine Rangueil, Toulouse 31062, France.
| |
Collapse
|
84
|
The Role of Radiation Therapy for Pancreatic Cancer in the Adjuvant and Neoadjuvant Settings. Surg Oncol Clin N Am 2017; 26:431-453. [PMID: 28576181 DOI: 10.1016/j.soc.2017.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is the third leading cause of cancer-related death in the United States. Although surgery remains the only curative treatment, chemotherapy and radiation therapy are frequently used. In the adjuvant setting, radiation is usually delivered with chemotherapy to eradicate residual microscopic or macroscopic disease in the resection bed. Neoadjuvant radiation therapy has become more frequently utilized. This article reviews the historical and modern literature regarding radiation therapy in the neoadjuvant and adjuvant settings, focusing on the evolution of radiation therapy techniques and clinical trials in an attempt to identify patients best suited to receiving radiation therapy.
Collapse
|
85
|
Bruno MJ. Interventional endoscopic ultrasonography: Where are we headed? Dig Endosc 2017; 29:503-511. [PMID: 28181708 DOI: 10.1111/den.12842] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/06/2017] [Indexed: 02/08/2023]
Abstract
Endoscopic ultrasonography (EUS) is an essential endoscopic tool within the diagnostic and therapeutic armamentarium of gastrointestinal and hepatic diseases. EUS-guided tissue acquisition will develop towards facilitating personalized treatment by obtaining large representative tissue specimens for elaborate immunohistochemical and biomolecular typing of the tumor. Intratumoral or intravascular delivery of drugs potentially offers many advantages over systemic injection. Intratumoral application of radiofrequency ablation and photodynamic therapy show promise but need to be explored further. Appositioning and connecting luminal structures within the gastrointestinal tract using fully covered expandable lumen-apposing stents will expand its indication far beyond the drainage of (infected) fluid collections and EUS-guided gastrojejunostomy is a particularly exciting development that could have significant impact on the management of gastric outlet obstruction.
Collapse
Affiliation(s)
- Marco J Bruno
- Department of Gastroenterology & Hepatology, Erasmus Medical Centre, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
86
|
Luthra AK, Mishra G. Novel diagnostic and therapeutic modalities using endoscopic ultrasound in pancreatic disease. GASTROINTESTINAL INTERVENTION 2017. [DOI: 10.18528/gii160036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Anjuli Kristin Luthra
- Section on General Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Girish Mishra
- Department of Gastroenterology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
87
|
Shukla V, Dalela M, Vij M, Weichselbaum R, Kharbanda S, Ganguli M, Kufe D, Singh H. Systemic delivery of the tumor necrosis factor gene to tumors by a novel dual DNA-nanocomplex in a nanoparticle system. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1833-1839. [PMID: 28343015 DOI: 10.1016/j.nano.2017.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/03/2017] [Accepted: 03/15/2017] [Indexed: 11/15/2022]
Abstract
Many cancers fail to respond to immunotherapy as a result of immune suppression by the tumor microenvironment. The exogenous expression of immune cytokines to reprogram the tumor microenvironment represents an approach to circumvent this suppression. The present studies describe the development of a novel dual nanoparticle (DNP) system for driving DNA expression vectors encoding inflammatory cytokines in tumor cells. The DNP system consists of a DNA expression vector-cationic peptide nanocomplex (NC) surrounded by a diblock polymeric NP. Tumor necrosis factor alpha (TNF) was selected as the prototype cytokine for this system, based on its pleotropic inflammatory and anti-cancer activities. Our results demonstrate that the DNP system is highly effective in driving expression of TNF in tumor cells. We also demonstrate that the DNPs are effective in inducing apoptosis and anti-tumor activity. These findings support a novel immunotherapeutic approach for the intratumoral delivery of DNA vectors that express inflammatory cytokines.
Collapse
Affiliation(s)
- Vasundhara Shukla
- Center for Biomedical Engineering, Indian Institute of Technology, Delhi, India
| | - Manu Dalela
- Center for Biomedical Engineering, Indian Institute of Technology, Delhi, India
| | - Manika Vij
- Institute of Genomics and Integrative Biology, Delhi, India
| | - Ralph Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL, USA
| | | | - Munia Ganguli
- Institute of Genomics and Integrative Biology, Delhi, India
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Harpal Singh
- Center for Biomedical Engineering, Indian Institute of Technology, Delhi, India; All India Institute of Medical Science, Delhi, India.
| |
Collapse
|
88
|
Signoretti M, Valente R, Repici A, Delle Fave G, Capurso G, Carrara S. Endoscopy-guided ablation of pancreatic lesions: Technical possibilities and clinical outlook. World J Gastrointest Endosc 2017; 9:41-54. [PMID: 28250896 PMCID: PMC5311472 DOI: 10.4253/wjge.v9.i2.41] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/12/2016] [Accepted: 12/09/2016] [Indexed: 02/05/2023] Open
Abstract
Endoscopic ultrasound (EUS) and endoscopic retrograde cholangiopancreatography (ERCP)-guided ablation procedures are emerging as a minimally invasive therapeutic alternative to radiological and surgical treatments for locally advanced pancreatic cancer (LAPC), pancreatic neuroendocrine tumours (PNETs), and pancreatic cystic lesions (PCLs). The advantages of treatment under endoscopic control are the real-time imaging guidance and the possibility to reach a deep target like the pancreas. Currently, radiofrequency probes specifically designed for ERCP or EUS ablation are available as well as hybrid cryotherm probe combining radiofrequency with cryotechnology. To date, many reports and case series have confirmed the safety and feasibility of that kind of ablation technique in the pancreatic setting. Moreover, EUS-guided fine-needle injection is emerging as a method to deliver ablative and anti-tumoral agents inside the tumuor. Ethanol injection has been proposed mostly for the treatment of PCLs and for symptomatic functioning PNETs, and the use of gemcitabine and paclitaxel is also interesting in this setting. EUS-guided injection of chemical or biological agents including mixed lymphocyte culture, oncolytic viruses, and immature dendritic cells has been investigated for the treatment of LAPC. Data on the long-term efficacy of these approaches, and large prospective randomized studies are needed to confirm the real clinical benefits of these techniques for the management of pancreatic lesions.
Collapse
|
89
|
Stereotactic body radiotherapy (SBRT) in recurrent or oligometastatic pancreatic cancer : A toxicity review of simultaneous integrated protection (SIP) versus conventional SBRT. Strahlenther Onkol 2017; 193:433-443. [PMID: 28138949 DOI: 10.1007/s00066-017-1099-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 01/06/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Stereotactic body radiotherapy (SBRT) in pancreatic cancer can be limited by its proximity to organs at risk (OAR). In this analysis, we evaluated the toxicity and efficacy of two different treatment approaches in patients with locally recurrent or oligometastatic pancreatic cancer. MATERIALS AND METHODS According to the prescription method, patients were divided in two cohorts (C1 and C2). The planning target volume (PTV) was created through a 4 mm expansion of the internal target volume. In C2, a subvolume was additionally created, a simultaneous integrated protection (SIP), which is the overlap of the PTV with the planning risk volume of an OAR to which we prescribed a reduced dose. RESULTS In all, 18 patients were treated (7 with local recurrences, 9 for oligometastases, 2 for both). Twelve of 23 lesions were treated without SIP (C1) and 11 with SIP (C2). The median follow-up was 12.8 months. Median overall survival (OS) was 13.2 (95% confidence interval [CI] 9.8-14.6) months. The OS rates at 6 and 12 months were 87 and 58%, respectively. Freedom from local progression for combined cohorts at 6 and 12 months was 93 and 67% (95% CI 15-36), respectively. Local control was not statistically different between the two groups. One patient in C2 experienced grade ≥3 acute toxicities and 1 patient in C1 experienced a grade ≥3 late toxicity. CONCLUSION The SIP approach is a useful prescription method for abdominal SBRT with a favorable toxicity profile which does not compromise local control and overall survival despite dose sacrifices in small subvolumes.
Collapse
|
90
|
Lee LS, Andersen DK, Ashida R, Brugge WR, Canto MI, Chang KJ, Chari ST, DeWitt J, Hwang JH, Khashab MA, Kim K, Levy MJ, McGrath K, Park WG, Singhi A, Stevens T, Thompson CC, Topazian MD, Wallace MB, Wani S, Waxman I, Yadav D, Singh VK. Endoscopic Ultrasound and Related Technologies for the Diagnosis and Treatment of Pancreatic Disease - Research Gaps and Opportunities: Summary of a National Institute of Diabetes and Digestive and Kidney Diseases Workshop. Pancreas 2017; 46:1242-1250. [PMID: 28926412 PMCID: PMC5645254 DOI: 10.1097/mpa.0000000000000936] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A workshop was sponsored by the National Institute of Diabetes and Digestive and Kidney Diseases to address the research gaps and opportunities in pancreatic endoscopic ultrasound (EUS). The event occurred on July 26, 2017 in 4 sessions: (1) benign pancreatic diseases, (2) high-risk pancreatic diseases, (3) diagnostic and therapeutics, and (4) new technologies. The current state of knowledge was reviewed, with identification of numerous gaps in knowledge and research needs. Common themes included the need for large multicenter consortia of various pancreatic diseases to facilitate meaningful research of these entities; to standardize EUS features of different pancreatic disorders, the technique of sampling pancreatic lesions, and the performance of various therapeutic EUS procedures; and to identify high-risk disease early at the cellular level before macroscopic disease develops. The need for specialized tools and accessories to enable the safe and effective performance of therapeutic EUS procedures also was discussed.
Collapse
Affiliation(s)
- Linda S. Lee
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Dana K. Andersen
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Reiko Ashida
- Departments of Cancer Survey and Gastrointestinal Oncology, Osaka Prefectural Hospital Organization, Osaka International Cancer Institute, Osaka, Japan
| | - William R. Brugge
- Department of Gastroenterology, Massachusetts General Hospital, Boston, MA
| | - Mimi I. Canto
- Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kenneth J. Chang
- Comprehensive Digestive Disease Center, Department of Gastroenterology and Hepatology, University of California at Irvine Health, Orange, CA
| | - Suresh T. Chari
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - John DeWitt
- Division of Gastroenterology, Indiana University Health Medical Center, Indianapolis, IN
| | - Joo Ha Hwang
- Department of Medicine, University of Washington, Seattle, WA
| | - Mouen A. Khashab
- Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kang Kim
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Michael J. Levy
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Kevin McGrath
- Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Walter G. Park
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Aatur Singhi
- Department of Pathology, University of Pittsburgh Medical Center, Sewickley, PA
| | - Tyler Stevens
- Department of Gastroenterology, Cleveland Clinic, Cleveland, OH
| | - Christopher C. Thompson
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Mark D. Topazian
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Michael B. Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic Florida, Jacksonville, FL
| | - Sachin Wani
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Irving Waxman
- Department of Medicine, The University of Chicago Comprehensive Cancer Center, University of Chicago School of Medicine, Chicago, IL
| | - Dhiraj Yadav
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Vikesh K. Singh
- Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
91
|
Yoo J, Kistler CA, Yan L, Dargan A, Siddiqui AA. Endoscopic ultrasound in pancreatic cancer: innovative applications beyond the basics. J Gastrointest Oncol 2016; 7:1019-1029. [PMID: 28078128 PMCID: PMC5177581 DOI: 10.21037/jgo.2016.08.07] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/06/2016] [Indexed: 12/16/2022] Open
Abstract
Endoscopic ultrasound (EUS) has become a mainstay in assisting in the diagnosis and staging of pancreatic cancer. In addition, EUS provides a modality to treat chronic pain through celiac plexus neurolysis. Currently, there is growing data and utilization of EUS in more diverse and innovative applications aimed at providing more sophisticated diagnostic, prognostic and therapeutic options for patients with pancreatic cancer. EUS delivery of chemotherapy, viral and biological vectors and fiducial markers may eventually revolutionize the way clinicians approach the care of a patient with pancreatic cancer.
Collapse
Affiliation(s)
- Joseph Yoo
- Department of Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - C. Andrew Kistler
- Department of Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
- Division of Gastroenterology and Hepatology, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Linda Yan
- Department of Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Andrew Dargan
- Department of Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Ali A. Siddiqui
- Department of Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
- Division of Gastroenterology and Hepatology, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| |
Collapse
|
92
|
Vassaux G, Angelova A, Baril P, Midoux P, Rommelaere J, Cordelier P. The Promise of Gene Therapy for Pancreatic Cancer. Hum Gene Ther 2016; 27:127-33. [PMID: 26603492 DOI: 10.1089/hum.2015.141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Unlike for other digestive cancer entities, chemotherapy, radiotherapy, and targeted therapies have, so far, largely failed to improve patient survival in pancreatic adenocarcinoma (PDAC), which remains the fourth leading cause of cancer-related death in Europe and the United States. In this context, gene therapy may offer a new avenue for patients with PDAC. In this review, we explore the research currently ongoing in French laboratories aimed at defeating PDAC using nonviral therapeutic gene delivery, targeted transgene expression, or oncolytic virotherapy that recently or will soon bridge the gap between experimental models of cancer and clinical trials. These studies are likely to change clinical practice or thinking about PDAC management, as they represent a major advance not only for PDAC but may also significantly influence the field of gene-based molecular treatment of cancer.
Collapse
Affiliation(s)
- Georges Vassaux
- 1 Université de Nice Sophia Antipolis , Nice, France .,2 Laboratoire TIRO , UMRE 4320, CEA, Nice, France
| | - Assia Angelova
- 3 German Cancer Research Center (DKFZ) , Tumor Virology/F010, Heidelberg, Germany
| | - Patrick Baril
- 4 Centre de Biophysique Moléculaire, CNRS UPR4301 and University of Orléans , Orléans, France
| | - Patrick Midoux
- 4 Centre de Biophysique Moléculaire, CNRS UPR4301 and University of Orléans , Orléans, France
| | - Jean Rommelaere
- 3 German Cancer Research Center (DKFZ) , Tumor Virology/F010, Heidelberg, Germany
| | - Pierre Cordelier
- 5 INSERM , UMR1037 CRCT, F-31000 Toulouse, France .,6 Université Toulouse III-Paul Sabatier , F-31000 Toulouse, France
| |
Collapse
|
93
|
Jamorabo DS, Lin SH, Jabbour SK. Successes and Failures of Combined Modalities in Upper Gastrointestinal Malignancies: New Directions. Semin Radiat Oncol 2016; 26:307-19. [PMID: 27619252 PMCID: PMC10794083 DOI: 10.1016/j.semradonc.2016.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Upper gastrointestinal malignancies generally have moderate to poor cure rates, even in the earliest stages, thereby implying that both local and systemic treatments have room for improvement. Therapeutic options are broadening, however, with the development of new immunotherapies and targeted agents, which can have synergistic effects with radiotherapy. Here we discuss the current state of combined modality therapy for upper gastrointestinal malignancies, specifically recent successes and setbacks in trials of radiation therapy with targeted therapies, vaccines, immunotherapies, and chemotherapies.
Collapse
Affiliation(s)
- Daniel S Jamorabo
- Department of Internal Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ.
| |
Collapse
|
94
|
Kang J, Demaria S, Formenti S. Current clinical trials testing the combination of immunotherapy with radiotherapy. J Immunother Cancer 2016; 4:51. [PMID: 27660705 PMCID: PMC5028964 DOI: 10.1186/s40425-016-0156-7] [Citation(s) in RCA: 295] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/02/2016] [Indexed: 01/12/2023] Open
Abstract
Increasing evidence demonstrates that radiation acts as an immune stimulus, recruiting immune mediators that enable anti-tumor responses within and outside the radiation field. There has been a rapid expansion in the number of clinical trials harnessing radiation to enhance antitumor immunity. If positive, results of these trials will lead to a paradigm shift in the use of radiotherapy. In this review, we discuss the rationale for trials combining radiation with various immunotherapies, provide an update of recent clinical trial results and highlight trials currently in progress. We also address issues pertaining to the optimal incorporation of immunotherapy with radiation, including sequencing of treatment, radiation dosing and evaluation of clinical trial endpoints.
Collapse
Affiliation(s)
- Josephine Kang
- Department of Radiation Oncology, Weill Cornell Medicine, 525 East 68th Street, New York, NY 10065 USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, 525 East 68th Street, New York, NY 10065 USA
| | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, 525 East 68th Street, New York, NY 10065 USA ; Department of Radiation Oncology, Stich Radiation Center, 525 East 68th Street, New York, NY 10065 USA
| |
Collapse
|
95
|
Coronel E, Waxman I. State-of-the-art endoscopic procedures for pancreatic cancer. Future Oncol 2016; 12:2037-47. [DOI: 10.2217/fon-2016-0123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is the twelfth most common cancer worldwide, taking the fourth place in cancer-related mortality in western countries. Despite significant efforts in understanding the tumor biology of pancreatic cancer and introducing new technologies and therapies to improve the detection, staging and treatment of this disease, pancreatic cancer continues to have a high and almost unchanged mortality. In the last few decades, the development of techniques such as endoscopic retrograde cholangio pancreatography and endoscopic ultrasound have allowed us to directly access the pancreaticobiliary system and fight pancreatic cancer and its complications from different fronts. Our goal with this review is to discuss the most cutting-edge endoscopic techniques available in our armamentarium to diagnose, stage and treat pancreatic cancer.
Collapse
Affiliation(s)
- Emmanuel Coronel
- University of Chicago Medicine, Gastroenterology, Hepatology & Nutrition, Chicago, IL, USA
| | - Irving Waxman
- University of Chicago Medicine, Center for Endoscopic Research & Therapeutics, Chicago IL, USA
| |
Collapse
|
96
|
Abstract
The human body combats infection and promotes wound healing through the remarkable process of inflammation. Inflammation is characterized by the recruitment of stromal cell activity including recruitment of immune cells and induction of angiogenesis. These cellular processes are regulated by a class of soluble molecules called cytokines. Based on function, cell target, and structure, cytokines are subdivided into several classes including: interleukins, chemokines, and lymphokines. While cytokines regulate normal physiological processes, chronic deregulation of cytokine expression and activity contributes to cancer in many ways. Gene polymorphisms of all types of cytokines are associated with risk of disease development. Deregulation RNA and protein expression of interleukins, chemokines, and lymphokines have been detected in many solid tumors and hematopoetic malignancies, correlating with poor patient prognosis. The current body of literature suggests that in some tumor types, interleukins and chemokines work against the human body by signaling to cancer cells and remodeling the local microenvironment to support the growth, survival, and invasion of primary tumors and enhance metastatic colonization. Some lymphokines are downregulated to suppress tumor progression by enhancing cytotoxic T cell activity and inhibiting tumor cell survival. In this review, we will describe the structure/function of several cytokine families and review our current understanding on the roles and mechanisms of cytokines in tumor progression. In addition, we will also discuss strategies for exploiting the expression and activity of cytokines in therapeutic intervention.
Collapse
Affiliation(s)
- M Yao
- University of Kansas Medical Center, Kansas City, KS, United States
| | - G Brummer
- University of Kansas Medical Center, Kansas City, KS, United States
| | - D Acevedo
- University of Kansas Medical Center, Kansas City, KS, United States
| | - N Cheng
- University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
97
|
Balaban EP, Mangu PB, Khorana AA, Shah MA, Mukherjee S, Crane CH, Javle MM, Eads JR, Allen P, Ko AH, Engebretson A, Herman JM, Strickler JH, Benson AB, Urba S, Yee NS. Locally Advanced, Unresectable Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2016; 34:2654-2668. [PMID: 27247216 DOI: 10.1200/jco.2016.67.5561] [Citation(s) in RCA: 268] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To provide evidence-based recommendations to oncologists and others for treatment of patients with locally advanced, unresectable pancreatic cancer. METHODS American Society of Clinical Oncology convened an Expert Panel of medical oncology, radiation oncology, surgical oncology, gastroenterology, palliative care, and advocacy experts and conducted a systematic review of the literature from January 2002 to June 2015. Outcomes included overall survival, disease-free survival, progression-free survival, and adverse events. RESULTS Twenty-six randomized controlled trials met the systematic review criteria. RECOMMENDATIONS A multiphase computed tomography scan of the chest, abdomen, and pelvis should be performed. Baseline performance status and comorbidity profile should be evaluated. The goals of care, patient preferences, psychological status, support systems, and symptoms should guide decisions for treatments. A palliative care referral should occur at first visit. Initial systemic chemotherapy (6 months) with a combination regimen is recommended for most patients (for some patients radiation therapy may be offered up front) with Eastern Cooperative Oncology Group performance status 0 or 1 and a favorable comorbidity profile. There is no clear evidence to support one regimen over another. The gemcitabine-based combinations and treatments recommended in the metastatic setting (eg, fluorouracil, leucovorin, irinotecan, and oxaliplatin and gemcitabine plus nanoparticle albumin-bound paclitaxel) have not been evaluated in randomized controlled trials involving locally advanced, unresectable pancreatic cancer. If there is local disease progression after induction chemotherapy, without metastasis, then radiation therapy or stereotactic body radiotherapy may be offered also with an Eastern Cooperative Oncology Group performance status ≤ 2 and an adequate comorbidity profile. If there is stable disease after 6 months of induction chemotherapy but unacceptable toxicities, radiation therapy may be offered as an alternative. Patients with disease progression should be offered treatment per the ASCO Metastatic Pancreatic Cancer Treatment Guideline. Follow-up visits every 3 to 4 months are recommended. Additional information is available at www.asco.org/guidelines/LAPC and www.asco.org/guidelines/MetPC and www.asco.org/guidelineswiki.
Collapse
Affiliation(s)
- Edward P Balaban
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Pamela B Mangu
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Alok A Khorana
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Manish A Shah
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Somnath Mukherjee
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Christopher H Crane
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Milind M Javle
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Jennifer R Eads
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Peter Allen
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Andrew H Ko
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Anitra Engebretson
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Joseph M Herman
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - John H Strickler
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Al B Benson
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Susan Urba
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| | - Nelson S Yee
- Edward P. Balaban, Cancer Care Partnership, State College; Edward P. Balaban and Nelson S. Yee, Penn State Hershey Cancer Institute, Hershey, PA; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Alok A. Khorana, Cleveland Clinic; Jennifer R. Eads, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH; Manish A. Shah, The Weill Cornell Medical Center; Peter Allen, Memorial Sloan Kettering Cancer Center, New York, NY; Somnath Mukherjee, University of Oxford, Oxford, United Kingdom; Christopher H. Crane and Milind M. Javle, The University of Texas MD Anderson Cancer Center, Houston, TX; Andrew H. Ko, University of California San Francisco Comprehensive Cancer Center, San Francisco, CA; Anitra Engebretson, Patient Representative, Portland, OR; Joseph M. Herman, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; John H. Strickler, Duke University Medical Center, Durham, NC; Al B. Benson III, Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL; and Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI
| |
Collapse
|
98
|
RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients. Oncotarget 2016; 6:24560-70. [PMID: 26009994 PMCID: PMC4695206 DOI: 10.18632/oncotarget.4183] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/02/2015] [Indexed: 12/12/2022] Open
Abstract
Purpose The miniature biodegradable implant siG12D-LODER™ was inserted into a tumor and released a siRNA drug against KRAS(G12D) along four months. This novel siRNA based drug was studied, in combination with chemotherapy, as targeted therapy for Locally Advanced Pancreatic Cancer (LAPC). Methods An open-label Phase 1/2a study in the first-line setting of patients with non-operable LAPC was initiated. In this study patients were assigned to receive a single dose of siG12D-LODERs, in three escalating dose cohorts (0.025mg, 0.75mg and 3.0mg). Gemcitabine was given on a weekly basis, following the siG12D-LODERTM insertion, until disease progression. The recommended dose was further examined with modified FOLFIRINOX. The follow up period was eight weeks and survival until death. Results Fifteen patients with LAPC were enrolled. Among the 15 treated patients, the most frequent adverse events observed were grade 1or 2 in severity (89%); five patients experienced serious adverse events (SAEs). In 12 patients analyzed by CT scans, none showed tumor progression, the majority (10/12) demonstrated stable disease and two showed partial response. Decrease in tumor marker CA19-9 was observed in 70% (7/10) of patients. Median overall survival was 15.12 months; 18 month survival was 38.5%. Conclusions The combination of siG12D-LODER™ and chemotherapy is well tolerated, safe and demonstrated a potential efficacy in patients with LAPC. NCT01188785
Collapse
|
99
|
Going viral: a review of replication-selective oncolytic adenoviruses. Oncotarget 2016; 6:19976-89. [PMID: 26280277 PMCID: PMC4652981 DOI: 10.18632/oncotarget.5116] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/09/2015] [Indexed: 12/12/2022] Open
Abstract
Oncolytic viruses have had a tumultuous course, from the initial anecdotal reports of patients having antineoplastic effects after natural viral infections a century ago to the development of current cutting-edge therapies in clinical trials. Adenoviruses have long been the workhorse of virotherapy, and we review both the scientific and the not-so-scientific forces that have shaped the development of these therapeutics from wild-type viral pathogens, turning an old foe into a new friend. After a brief review of the mechanics of viral replication and how it has been modified to engineer tumor selectivity, we give particular attention to ONYX-015, the forerunner of virotherapy with extensive clinical testing that pioneered the field. The findings from those as well as other oncolytic trials have shaped how we now view these viruses, which our immune system has evolved to vigorously attack, as promising immunotherapy agents.
Collapse
|
100
|
Biopharmaceuticals and gene vectors opening new avenues in cancer immune therapy. Ther Deliv 2016; 7:419-22. [DOI: 10.4155/tde-2016-0032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|