51
|
Li S, Xu HX, Wu CT, Wang WQ, Jin W, Gao HL, Li H, Zhang SR, Xu JZ, Qi ZH, Ni QX, Yu XJ, Liu L. Angiogenesis in pancreatic cancer: current research status and clinical implications. Angiogenesis 2018; 22:15-36. [PMID: 30168025 DOI: 10.1007/s10456-018-9645-2] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 08/20/2018] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer is one of the most lethal malignancies worldwide. Although the standard of care in pancreatic cancer has improved, prognoses for patients remain poor with a 5-year survival rate of < 5%. Angiogenesis, namely, the formation of new blood vessels from pre-existing vessels, is an important event in tumor growth and hematogenous metastasis. It is a dynamic and complex process involving multiple mechanisms and is regulated by various molecules. Inhibition of angiogenesis has been an established therapeutic strategy for many solid tumors. However, clinical outcomes are far from satisfying for pancreatic cancer patients receiving anti-angiogenic therapies. In this review, we summarize the current status of angiogenesis in pancreatic cancer research and explore the reasons for the poor efficacy of anti-angiogenic therapies, aiming to identify some potential therapeutic targets that may enhance the effectiveness of anti-angiogenic treatments.
Collapse
Affiliation(s)
- Shuo Li
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Hua-Xiang Xu
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chun-Tao Wu
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Wen-Quan Wang
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Wei Jin
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - He-Li Gao
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Hao Li
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Shi-Rong Zhang
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jin-Zhi Xu
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zi-Hao Qi
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Quan-Xing Ni
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xian-Jun Yu
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Liang Liu
- Department of Pancreatic & Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong An Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
52
|
Chin V, Nagrial A, Sjoquist K, O'Connor CA, Chantrill L, Biankin AV, Scholten RJPM, Yip D. 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: 56] [Impact Index Per Article: 8.0] [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
|
53
|
Rosenberg A, Mahalingam D. Immunotherapy in pancreatic adenocarcinoma-overcoming barriers to response. J Gastrointest Oncol 2018; 9:143-159. [PMID: 29564181 PMCID: PMC5848027 DOI: 10.21037/jgo.2018.01.13] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 12/28/2017] [Indexed: 12/12/2022] Open
Abstract
Pancreatic adenocarcinoma (PAC) remains one of the leading causes of cancer-related death. Despite multiple advances in targeted and immune therapies, the 5-year survival in advanced PAC remains poor. In this review, we discuss some of the unique aspects of the tumor microenvironment (TME) in PAC that may contribute to its resistance to immune therapies, as well as opportunities to potentially overcome some of these inherent barriers. Furthermore, we discuss strategies to enable immune therapies in PAC such as cytotoxic chemotherapy and radiation therapy, cancer vaccines, cytokine based therapy, oncolytic viruses, and adoptive T-cell therapy. Finally, we address a variety of targeted therapies as a strategy to further amplify immune responses in PAC.
Collapse
Affiliation(s)
- Ari Rosenberg
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | | |
Collapse
|
54
|
Nishida S, Ishikawa T, Egawa S, Koido S, Yanagimoto H, Ishii J, Kanno Y, Kokura S, Yasuda H, Oba MS, Sato M, Morimoto S, Fujiki F, Eguchi H, Nagano H, Kumanogoh A, Unno M, Kon M, Shimada H, Ito K, Homma S, Oka Y, Morita S, Sugiyama H. Combination Gemcitabine and WT1 Peptide Vaccination Improves Progression-Free Survival in Advanced Pancreatic Ductal Adenocarcinoma: A Phase II Randomized Study. Cancer Immunol Res 2018; 6:320-331. [PMID: 29358173 DOI: 10.1158/2326-6066.cir-17-0386] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/17/2017] [Accepted: 01/09/2018] [Indexed: 11/16/2022]
Abstract
We investigated the efficacy of a Wilms' tumor gene 1 (WT1) vaccine combined with gemcitabine (GEMWT1) and compared it with gemcitabine (GEM) monotherapy for advanced pancreatic ductal adenocarcinoma (PDAC) in a randomized phase II study. We randomly assigned HLA-A*02:01- or HLA-A*24:02-positive patients with advanced PDAC to receive GEMWT1 or GEM. We assessed WT1-specific immune responses via delayed-type hypersensitivity (DTH) to the WT1 peptide and a tetramer assay to detect WT1-specific cytotoxic T lymphocytes (WT1-CTL). Of 91 patients enrolled, 85 were evaluable (GEMWT1: n = 42; GEM: n = 43). GEMWT1 prolonged progression-free survival [PFS; hazard ratio (HR), 0.66; P = 0.084] and improved overall survival rate at 1 year (1-year OS%; GEMWT1: 35.7%; GEM: 20.9%). However, the difference in OS was not significant (HR: 0.82; P = 0.363). These effects were particularly evident in metastatic PDAC (PFS: HR 0.51, P = 0.0017; 1-year OS%: GEMWT1 27.3%; GEM 11.8%). The combination was well tolerated, with no unexpected serious adverse events. In patients with metastatic PDAC, PFS in the DTH-positive GEMWT1 group was significantly prolonged, with a better HR of 0.27 compared with the GEM group, whereas PFS in the DTH-negative GEMWT1 group was similar to that in the GEM group (HR 0.86; P = 0.001). DTH positivity was associated with an increase in WT1-CTLs induced by the WT1 vaccine. GEM plus the WT1 vaccine prolonged PFS and may improve 1-year OS% in advanced PDAC. These clinical effects were associated with the induction of WT1-specific immune responses. Cancer Immunol Res; 6(3); 320-31. ©2018 AACR.
Collapse
Affiliation(s)
- Sumiyuki Nishida
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Takeshi Ishikawa
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shinichi Egawa
- Division of International Cooperation for Disaster Medicine, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Shigeo Koido
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Jun Ishii
- Division of General and Gastroenterological Surgery, Department of Surgery, Toho University Faculty of Medicine, Tokyo, Japan
| | - Yoshihide Kanno
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Satoshi Kokura
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroaki Yasuda
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mari Saito Oba
- Department of Biostatistics, Yokohama City University, Yokohama, Japan
| | - Maho Sato
- Department of Biostatistics, Yokohama City University, Yokohama, Japan
| | - Soyoko Morimoto
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masanori Kon
- Department of Surgery, Kansai Medical University, Hirakata, Japan
| | - Hideaki Shimada
- Division of General and Gastroenterological Surgery, Department of Surgery, Toho University Faculty of Medicine, Tokyo, Japan
| | - Kei Ito
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Sadamu Homma
- Division of Oncology, Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshihiro Oka
- Department of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
55
|
Schmitz-Winnenthal FH, Hohmann N, Schmidt T, Podola L, Friedrich T, Lubenau H, Springer M, Wieckowski S, Breiner KM, Mikus G, Büchler MW, Keller AV, Koc R, Springfeld C, Knebel P, Bucur M, Grenacher L, Haefeli WE, Beckhove P. A phase 1 trial extension to assess immunologic efficacy and safety of prime-boost vaccination with VXM01, an oral T cell vaccine against VEGFR2, in patients with advanced pancreatic cancer. Oncoimmunology 2018; 7:e1303584. [PMID: 29632710 DOI: 10.1080/2162402x.2017.1303584] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/24/2017] [Accepted: 03/01/2017] [Indexed: 12/17/2022] Open
Abstract
VXM01 is a first-in-kind orally applied tumor vaccine based on live attenuated Salmonella typhi carrying an expression plasmid encoding VEGFR2, an antigen expressed on tumor vasculature and a stable and accessible target for anti-angiogenic intervention. A recent randomized, placebo-controlled, phase I dose-escalation trial in advanced pancreatic cancer patients demonstrated safety, immunogenicity and transient, T-cell response-related anti-angiogenic activity of four priming vaccinations applied within one week. We here evaluated whether monthly boost vaccinations are safe and can sustain increased frequencies of vaccine-specific T cells. Patients with advanced pancreatic cancer were randomly assigned at a ratio of 2:1 to priming with VXM01 followed by up to six monthly boost vaccinations, or placebo treatment. Vaccinations were applied orally at two alternative doses of either 106 colony-forming units (CFU) or 107 CFU, and concomitant treatment with standard-of-care gemcitabine during the priming phase, and any treatment thereafter, was allowed in the study. Immunomonitoring involved interferon-gamma (IFNγ) ELIspot analysis with long overlapping peptides spanning the entire VEGFR2 sequence. A total of 26 patients were treated. Treatment-related adverse events preferentially associated with VXM01 were decreases in lymphocyte numbers in the blood, increased frequencies of neutrophils and diarrhea. Eight out of 16 patients who received at least one boosting vaccination responded with pronounced, i.e. at least 3-fold, increase in VEGFR2-specific T cell response over baseline levels. In the VXM01 vaccination group, VEGFR2-specific T cells peaked preferentially during the boosting phase with an average 4-fold increase over baseline levels. In conclusion, prime/boost vaccination with VXM01 was safe and immunogenic and increased vaccine specific T cell responses compared with placebo treatment.
Collapse
Affiliation(s)
| | - Nicolas Hohmann
- Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Schmidt
- Department of General, Abdominal and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Lilli Podola
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany.,Medical Oncology, National Center for Tumor Diseases, Heidelberg, Germany
| | - Tobias Friedrich
- Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | | | | | | | | | - Gerd Mikus
- Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus W Büchler
- Department of General, Abdominal and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Ruhan Koc
- Department of General, Abdominal and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Phillip Knebel
- Department of General, Abdominal and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Mariana Bucur
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Lars Grenacher
- Diagnostic Munich, Diagnostic Prevention and Imaging Center, Munich, Germany
| | - Walter E Haefeli
- Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp Beckhove
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany.,Medical Oncology, National Center for Tumor Diseases, Heidelberg, Germany
| |
Collapse
|
56
|
Beatty GL, Eghbali S, Kim R. Deploying Immunotherapy in Pancreatic Cancer: Defining Mechanisms of Response and Resistance. Am Soc Clin Oncol Educ Book 2017; 37:267-278. [PMID: 28561678 DOI: 10.1200/edbk_175232] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The immune reaction to pancreatic ductal adenocarcinoma (PDAC) is a strong prognostic determinant of clinical outcomes and may be a promising therapeutic target. We use multiplex immunohistochemistry to illustrate distinct patterns of T-cell and myeloid cell infiltration seen in PDAC that have therapeutic implications and discuss the current state of immunotherapy in this disease. Based on collective findings from clinical and preclinical studies, two conceptual models have emerged for applying immunotherapy in PDAC that involve (1) restoring elements of T-cell immunosurveillance and (2) redirecting myeloid cells to condition tumors with increased sensitivity to cytotoxic therapies. Overall, the success of immunotherapy in PDAC will most likely rely on strategic combinations of therapies that are informed by well-designed correlative analyses that consider the spatial heterogeneity of immune responses detected in malignant tissues.
Collapse
Affiliation(s)
- Gregory L Beatty
- From the Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA; Department of Medicine, Division of Hematology-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Shabnam Eghbali
- From the Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA; Department of Medicine, Division of Hematology-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Rebecca Kim
- From the Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA; Department of Medicine, Division of Hematology-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
57
|
Wada S, Yada E, Ohtake J, Sasada T. Personalized peptide vaccines for cancer therapy: current progress and state of the art. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2017. [DOI: 10.1080/23808993.2017.1403286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Satoshi Wada
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| | - Erica Yada
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| | - Junya Ohtake
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| | - Tetsuro Sasada
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| |
Collapse
|
58
|
Jackson DO, Byrd K, Vreeland TJ, Hale DF, Herbert GS, Greene JM, Schneble EJ, Berry JS, Trappey AF, Clifton GT, Hardin MO, Martin J, Elkas JC, Conrads TP, Darcy KM, Hamilton CA, Maxwell GL, Peoples GE. Interim analysis of a phase I/IIa trial assessing E39+GM-CSF, a folate binding protein vaccine, to prevent recurrence in ovarian and endometrial cancer patients. Oncotarget 2017; 8:15912-15923. [PMID: 27852036 PMCID: PMC5362533 DOI: 10.18632/oncotarget.13305] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/19/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Folate binding protein(FBP) is an immunogenic protein over-expressed in endometrial(EC) and ovarian cancer(OC). We are conducting a phase I/IIa trial of E39 (GALE 301)+GM-CSF, an HLA-A2-restricted, FBP-derived peptide vaccine to prevent recurrences in disease-free EC and OC patients. This interim analysis summarizes toxicity, immunologic responses, and clinical outcomes to date. METHODS HLA-A2+ patients were vaccinated(VG), and HLA-A2- or -A2+ patients were followed as controls(CG). Six monthly intradermal inoculations of E39+250mcg GM-CSF were administered to VG. Demographic, safety, immunologic, and recurrence rate(RR) data were collected and evaluated. RESULTS This trial enrolled 51 patients; 29 in the VG and 22 in the CG. Fifteen patients received 1000mcg E39, and 14 received <1000mcg. There were no clinicopathologic differences between groups(all p ≥ 0.1). E39 was well-tolerated regardless of dose. DTH increased pre- to post-vaccination (5.7±1.5 mm vs 10.3±3.0 mm, p = 0.06) in the VG, and increased more in the 1000mcg group (3.8±2.0 mm vs 9.5±3.5 mm, p = 0.03). With 12 months median follow-up, the RR was 41% (VG) vs 55% (CG), p = 0.41. Among the 1000mcg patients, the RR was 13.3% vs 55% CG, p = 0.01. Estimated 2-year DFS was 85.7% in the 1000mcg group vs 33.6% in the CG (p = 0.021). CONCLUSIONS This phase I/IIa trial reveals that E39+GM-CSF is well-tolerated and elicits a strong, dose-dependent in vivo immune response. Early efficacy results are promising in the 1000 mcg dose cohort. This study proves the safety and establishes the dose of E39 for a larger prospective, randomized, controlled trial in HLA-A2+ EC and OC patients to prevent recurrence.
Collapse
Affiliation(s)
- Doreen O Jackson
- Department of Surgery, San Antonio Military Medical Center, San Antonio, TX, USA
| | - Kevin Byrd
- National Capital Consortium Fellowship in Gynecologic Oncology, Walter Reed National Military Medical Center, Bethesda, MD, USA.,Gynecologic Cancer Center of Excellence, Annandale, VA, USA
| | - Timothy J Vreeland
- Department of Surgery, Womack Army Medical Center, Fayetteville, NC, USA
| | - Diane F Hale
- Department of Surgery, San Antonio Military Medical Center, San Antonio, TX, USA
| | - Garth S Herbert
- Department of Surgery, San Antonio Military Medical Center, San Antonio, TX, USA
| | - Julia M Greene
- Department of Surgery, San Antonio Military Medical Center, San Antonio, TX, USA
| | - Erika J Schneble
- Department of Surgery, San Antonio Military Medical Center, San Antonio, TX, USA
| | - John S Berry
- Department of Surgery, San Antonio Military Medical Center, San Antonio, TX, USA
| | - Alfred F Trappey
- Department of Surgery, San Antonio Military Medical Center, San Antonio, TX, USA
| | - G T Clifton
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark O Hardin
- Department of Surgery, Madigan Army Medical Center, Tacoma, WA, USA
| | | | - John C Elkas
- Department of Obstetrics and Gynecology, Inova Fairfax Hospital, Annandale, VA, USA.,Mid-Atlantic Gynecologic Oncology and Pelvic Surgical Associates, Annandale, VA, USA
| | - Thomas P Conrads
- National Capital Consortium Fellowship in Gynecologic Oncology, Walter Reed National Military Medical Center, Bethesda, MD, USA.,Gynecologic Cancer Center of Excellence, Annandale, VA, USA.,Department of Obstetrics and Gynecology, Inova Fairfax Hospital, Annandale, VA, USA.,Inova Schar Cancer Institute, Inova Health System, Annandale, VA, USA
| | - Kathleen M Darcy
- National Capital Consortium Fellowship in Gynecologic Oncology, Walter Reed National Military Medical Center, Bethesda, MD, USA.,Gynecologic Cancer Center of Excellence, Annandale, VA, USA
| | - Chad A Hamilton
- National Capital Consortium Fellowship in Gynecologic Oncology, Walter Reed National Military Medical Center, Bethesda, MD, USA.,Gynecologic Cancer Center of Excellence, Annandale, VA, USA
| | - George L Maxwell
- National Capital Consortium Fellowship in Gynecologic Oncology, Walter Reed National Military Medical Center, Bethesda, MD, USA.,Gynecologic Cancer Center of Excellence, Annandale, VA, USA.,Department of Obstetrics and Gynecology, Inova Fairfax Hospital Annandale, VA, USA.,Inova Schar Cancer Institute, Inova Health System, Annandale, VA, USA
| | | |
Collapse
|
59
|
Ottaiano A, Capozzi M, De Divitiis C, De Stefano A, Botti G, Avallone A, Tafuto S. Gemcitabine mono-therapy versus gemcitabine plus targeted therapy in advanced pancreatic cancer: a meta-analysis of randomized phase III trials. Acta Oncol 2017; 56:377-383. [PMID: 28256961 DOI: 10.1080/0284186x.2017.1288922] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Prognosis of advanced pancreatic cancer is dismal and the novel targeted therapies, albeit successfully used to treat many advanced tumors, have shown modest results. We performed a meta-analysis in order to quantify the effect size on survival of adding targeted therapy to single agent gemcitabine. METHODS Randomized phase III trials comparing gemcitabine mono-therapy versus gemcitabine plus a targeted agent in first-line treatment of advanced pancreatic cancer designed on survival as primary outcome were selected. Search was done through Medline and the registry of the NIH. Keywords used for searching were 'pancreas', 'pancreatic', 'gemcitabine'. Study quality was assessed with MERGE criteria. Findings were depicted in classical Forest plots. Publication bias was evaluated by the construction of funnel plot. RESULTS Nine studies met the meta-analysis inclusion criteria including 4564 patients. The target therapies were: erlotinib, cetuximab, rigosertib, elpamotide, bevacizumab, aflibercept, axitinib, masitinib and ganitumab. There was no statistically significant heterogeneity among the nine trials (p = 0.77). The hazard ratio (HR) of the pooled analysis was 0.998 (CI 95%: 0.932-1.068). Subgroup meta-analysis was also performed in anti-EGFR and anti-angiogenesis trials: the pooled HR were 0.94 (CI 95%: 0.705-1.175) and 1.055 (CI 95%: 0.913-1.197), respectively. CONCLUSIONS The present meta-analysis does not show significant improvements in survival for targeted drugs in advanced pancreatic cancer. The possible reason of these results could be linked to the biology of pancreatic cancer as well as to the absence of predictive factors.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- Department of Abdominal Oncology, Istituto Nazionale Tumori di Napoli “G. Pascale” IRCCS, National Cancer Institute, Naples, Italy
| | - Monica Capozzi
- Department of Abdominal Oncology, Istituto Nazionale Tumori di Napoli “G. Pascale” IRCCS, National Cancer Institute, Naples, Italy
| | - Chiara De Divitiis
- Department of Abdominal Oncology, Istituto Nazionale Tumori di Napoli “G. Pascale” IRCCS, National Cancer Institute, Naples, Italy
| | - Alfonso De Stefano
- Department of Abdominal Oncology, Istituto Nazionale Tumori di Napoli “G. Pascale” IRCCS, National Cancer Institute, Naples, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori di Napoli “G. Pascale” IRCCS, National Cancer Institute, Naples, Italy
| | - Antonio Avallone
- Department of Abdominal Oncology, Istituto Nazionale Tumori di Napoli “G. Pascale” IRCCS, National Cancer Institute, Naples, Italy
| | - Salvatore Tafuto
- Department of Abdominal Oncology, Istituto Nazionale Tumori di Napoli “G. Pascale” IRCCS, National Cancer Institute, Naples, Italy
| |
Collapse
|
60
|
Miyazawa M, Katsuda M, Maguchi H, Katanuma A, Ishii H, Ozaka M, Yamao K, Imaoka H, Kawai M, Hirono S, Okada KI, Yamaue H. Phase II clinical trial using novel peptide cocktail vaccine as a postoperative adjuvant treatment for surgically resected pancreatic cancer patients. Int J Cancer 2017; 140:973-982. [PMID: 27861852 DOI: 10.1002/ijc.30510] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 10/24/2016] [Indexed: 12/18/2022]
Abstract
We investigated peptide cocktail vaccine OCV-C01 containing epitope peptides derived from KIF20A, vascular endothelial growth factor receptor (VEGFR)1 and VEGFR2 combined with gemcitabine in the adjuvant treatment for resected pancreatic cancer patients. A single-arm multicenter phase II study was performed on 30 patients with pancreatic ductal carcinoma who underwent pancreatectomy. At each 28-day treatment cycle, patients received weekly subcutaneous injection of OCV-C01 for 48 weeks and gemcitabine was administered intravenously at 1,000 mg/m2 on days 1, 8 and 15 for 24 weeks. Patients were followed for 18 months. The primary endpoint was disease-free survival (DFS) and secondary endpoints included safety, overall survival (OS) and immunological assays on peptide-specific cytotoxic T lymphocyte (CTL) activity and KIF20A expression in resected pancreatic cancer. The median DFS was 15.8 months [95% confidence interval (CI), 11.1-20.6] and the DFS rate at 18 months was 34.6% (95% CI, 18.3-51.6). The median OS was not reached and the OS rate at 18 months was 69.0% (95% CI, 48.8-82.5). The administration of OCV-C01 was well tolerated. In the per protocol set, there were significant differences in DFS between patients with KIF20A-specific CTL responses and without (p = 0.027), and between patients with KIF20A expression and without (p = 0.014). In addition, all four patients who underwent R0 resection with KIF20A expression had no recurrence of pancreatic cancer with KIF20A-specific CTL responses. OCV-C01 combined with gemcitabine was tolerable with a median DFS of 15.8 months, which was favorable compared with previous data for resected pancreatic cancer.
Collapse
Affiliation(s)
- Motoki Miyazawa
- Second Department of Surgery, Wakayama Medical University School of Medicine, Japan
| | - Masahiro Katsuda
- Second Department of Surgery, Wakayama Medical University School of Medicine, Japan
| | | | - Akio Katanuma
- Center for Gastroenterology, Teine-Keijinkai Hospital, Japan
| | - Hiroshi Ishii
- Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Japan
| | - Masato Ozaka
- Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Japan
| | - Kenji Yamao
- Department of Gastroenterology, Aichi Cancer Center Hospital, Japan
| | - Hiroshi Imaoka
- Department of Gastroenterology, Aichi Cancer Center Hospital, Japan
| | - Manabu Kawai
- Second Department of Surgery, Wakayama Medical University School of Medicine, Japan
| | - Seiko Hirono
- Second Department of Surgery, Wakayama Medical University School of Medicine, Japan
| | - Ken-Ichi Okada
- Second Department of Surgery, Wakayama Medical University School of Medicine, Japan
| | - Hiroki Yamaue
- Second Department of Surgery, Wakayama Medical University School of Medicine, Japan
| |
Collapse
|
61
|
Kyrochristos ID, Glantzounis GK, Ziogas DE, Gizas I, Schizas D, Lykoudis EG, Felekouras E, Machairas A, Katsios C, Liakakos T, Cho WC, Roukos DH. From Clinical Standards to Translating Next-Generation Sequencing Research into Patient Care Improvement for Hepatobiliary and Pancreatic Cancers. Int J Mol Sci 2017; 18:E180. [PMID: 28106782 PMCID: PMC5297812 DOI: 10.3390/ijms18010180] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 12/19/2016] [Accepted: 12/27/2016] [Indexed: 02/06/2023] Open
Abstract
Hepatobiliary and pancreatic (HBP) cancers are associated with high cancer-related death rates. Surgery aiming for complete tumor resection (R0) remains the cornerstone of the treatment for HBP cancers. The current progress in the adjuvant treatment is quite slow, with gemcitabine chemotherapy available only for pancreatic ductal adenocarcinoma (PDA). In the advanced and metastatic setting, only two targeted drugs have been approved by the Food & Drug Administration (FDA), which are sorafenib for hepatocellular carcinoma and erlotinib for PDA. It is a pity that multiple Phase III randomized control trials testing the efficacy of targeted agents have negative results. Failure in the development of effective drugs probably reflects the poor understanding of genome-wide alterations and molecular mechanisms orchestrating therapeutic resistance and recurrence. In the post-ENCODE (Encyclopedia of DNA Elements) era, cancer is referred to as a highly heterogeneous and systemic disease of the genome. The unprecedented potential of next-generation sequencing (NGS) technologies to accurately identify genetic and genomic variations has attracted major research and clinical interest. The applications of NGS include targeted NGS with potential clinical implications, while whole-exome and whole-genome sequencing focus on the discovery of both novel cancer driver genes and therapeutic targets. These advances dictate new designs for clinical trials to validate biomarkers and drugs. This review discusses the findings of available NGS studies on HBP cancers and the limitations of genome sequencing analysis to translate genome-based biomarkers and drugs into patient care in the clinic.
Collapse
Affiliation(s)
- Ioannis D Kyrochristos
- Centre for Biosystems and Genome Network Medicine, Ioannina University, 45110 Ioannina, Greece.
- Department of Surgery, Ioannina University Hospital, 45110 Ioannina, Greece.
| | | | - Demosthenes E Ziogas
- Centre for Biosystems and Genome Network Medicine, Ioannina University, 45110 Ioannina, Greece.
- Department of Surgery, 'G. Hatzikosta' General Hospital, 45001 Ioannina, Greece.
| | | | - Dimitrios Schizas
- 1st Department of Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Efstathios G Lykoudis
- Department of Plastic Surgery, Ioannina University School of Medicine, 45110 Ioannina, Greece.
| | - Evangelos Felekouras
- 1st Department of Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Anastasios Machairas
- Third Department of Surgery, Attikon General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece.
| | - Christos Katsios
- Department of Surgery, Ioannina University Hospital, 45110 Ioannina, Greece.
| | - Theodoros Liakakos
- 1st Department of Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China.
| | - Dimitrios H Roukos
- Centre for Biosystems and Genome Network Medicine, Ioannina University, 45110 Ioannina, Greece.
- Department of Surgery, Ioannina University Hospital, 45110 Ioannina, Greece.
- Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece.
| |
Collapse
|
62
|
Suzuki N, Hazama S, Iguchi H, Uesugi K, Tanaka H, Hirakawa K, Aruga A, Hatori T, Ishizaki H, Umeda Y, Fujiwara T, Ikemoto T, Shimada M, Yoshimatsu K, Shimizu R, Hayashi H, Sakata K, Takenouchi H, Matsui H, Shindo Y, Iida M, Koki Y, Arima H, Furukawa H, Ueno T, Yoshino S, Nakamura Y, Oka M, Nagano H. Phase II clinical trial of peptide cocktail therapy for patients with advanced pancreatic cancer: VENUS-PC study. Cancer Sci 2017; 108:73-80. [PMID: 27783849 PMCID: PMC5276830 DOI: 10.1111/cas.13113] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 10/12/2016] [Accepted: 10/24/2016] [Indexed: 12/15/2022] Open
Abstract
We previously conducted a phase I clinical trial combining the HLA-A*2402-restricted KIF20A-derived peptide vaccine with gemcitabine for advanced pancreatic cancer (PC) and confirmed its safety and immunogenicity in cancer patients. In this study, we conducted a multicenter, single-armed, phase II trial using two antiangiogenic cancer vaccines targeting VEGFR1 and VEGFR2 in addition to the KIF20A peptide. We attempted to evaluate the clinical benefit of the cancer vaccination in combination with gemcitabine. Chemotherapy naïve PC patients were enrolled to evaluate primarily the 1-year survival rate, and secondarily overall survival (OS), progression free survival (PFS), response rate (RR), disease control rate (DCR) and the peptide-specific immune responses. All enrolled patients received therapy without the HLA-A information, and the HLA genotypes were used for classification of the patients. Between June 2012 and May 2013, a total of 68 patients were enrolled. No severe systemic adverse effects of Grade 3 or higher related to these three peptides were observed. The 1-year survival rates between the HLA-A*2402-matched and -unmatched groups were not significantly different. In the HLA-A*2402 matched group, patients showing peptide-specific CTL induction for KIF20A or VEGFR1 showed a better prognosis compared to those without such induction (P = 0.023, P = 0.009, respectively). In the HLA-A*2402-matched group, the patients who showed a strong injection site reaction had a better survival rate (P = 0.017) compared to those with a weak or no injection site reaction. This phase II study demonstrated that this therapeutic peptide cocktail might be effective in patients who demonstrate peptide-specific immune reactions although predictive biomarkers are needed for patient selection in its further clinical application.
Collapse
Affiliation(s)
- Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Shoichi Hazama
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Haruo Iguchi
- Clinical Research CenterShikoku Cancer Center, NHOMatsuyamaJapan
| | - Kazuhiro Uesugi
- Clinical Research CenterShikoku Cancer Center, NHOMatsuyamaJapan
| | - Hiroaki Tanaka
- Department of Surgical OncologyOsaka City University Graduate School of MedicineOsakaJapan
| | - Kosei Hirakawa
- Department of Surgical OncologyOsaka City University Graduate School of MedicineOsakaJapan
| | - Atsushi Aruga
- Institute of GastroenterologyTokyo Women's Medical UniversityTokyoJapan
| | - Takashi Hatori
- Institute of GastroenterologyTokyo Women's Medical UniversityTokyoJapan
| | - Hidenobu Ishizaki
- Department of Surgical Oncology and Regulation of Organ FunctionMiyazaki University School of MedicineMiyazakiJapan
| | - Yuzo Umeda
- Department of Gastroenterological SurgeryOkayama University Graduate School of MedicineOkayamaJapan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological SurgeryOkayama University Graduate School of MedicineOkayamaJapan
| | - Tetsuya Ikemoto
- Department of Digestive and Transplant SurgeryTokushima University Graduate School of MedicineTokushimaJapan
| | - Mitsuo Shimada
- Department of Digestive and Transplant SurgeryTokushima University Graduate School of MedicineTokushimaJapan
| | - Kazuhiko Yoshimatsu
- Department of SurgeryTokyo Women's Medical University Medical Center EastTokyoJapan
| | - Ryoichi Shimizu
- Department of SurgeryOgori Dai‐ichi General HospitalYamaguchiJapan
| | - Hiroto Hayashi
- Department of SurgeryKanmon Medical Center, NHOShimonosekiJapan
| | - Koichiro Sakata
- Department of SurgeryShimonoseki Medical Center, JCHOShimonosekiJapan
| | - Hiroko Takenouchi
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Hiroto Matsui
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Yoshitaro Shindo
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Michihisa Iida
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Yasunobu Koki
- Department of PharmacyYamaguchi University HospitalUbeJapan
| | - Hideki Arima
- Department of PharmacyYamaguchi University HospitalUbeJapan
| | | | - Tomio Ueno
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Shigefumi Yoshino
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Yusuke Nakamura
- Section of Hematology/OncologyDepartment of MedicineThe University of ChicagoChicagoIllinoisUSA
| | | | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| |
Collapse
|
63
|
Wada S, Yada E, Ohtake J, Fujimoto Y, Uchiyama H, Yoshida S, Sasada T. Current status and future prospects of peptide-based cancer vaccines. Immunotherapy 2016; 8:1321-1333. [PMID: 27993087 DOI: 10.2217/imt-2016-0063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cancer immunotherapy has attracted attention worldwide owing to the recent development of immune checkpoint inhibitors. However, these therapies have shown limited efficacy, and further advancements are needed before these modalities can progress to widespread use. Immune checkpoint inhibitors are a type of nonspecific cancer immunotherapy, and antitumor effects are only observed when cancer-specific T cells are found within the nonspecifically activated T-cell group. In order to facilitate the development of potent immunotherapies, selective enhancement of cancer-specific T cells is essential. In this report, we discuss current and future perspectives, including the latest clinical trials of cancer-specific immunotherapies, particularly cancer peptide vaccines.
Collapse
Affiliation(s)
- Satoshi Wada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao Asahi-ku, Yokohama, Kanagawa 241-8515, Japan
| | - Erika Yada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao Asahi-ku, Yokohama, Kanagawa 241-8515, Japan
| | - Junya Ohtake
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao Asahi-ku, Yokohama, Kanagawa 241-8515, Japan
| | - Yuki Fujimoto
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao Asahi-ku, Yokohama, Kanagawa 241-8515, Japan
| | - Hidemi Uchiyama
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao Asahi-ku, Yokohama, Kanagawa 241-8515, Japan
| | - Shintaro Yoshida
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao Asahi-ku, Yokohama, Kanagawa 241-8515, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao Asahi-ku, Yokohama, Kanagawa 241-8515, Japan
| |
Collapse
|
64
|
Silva APS, Coelho PV, Anazetti M, Simioni PU. Targeted therapies for the treatment of non-small-cell lung cancer: Monoclonal antibodies and biological inhibitors. Hum Vaccin Immunother 2016; 13:843-853. [PMID: 27831000 PMCID: PMC5404364 DOI: 10.1080/21645515.2016.1249551] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The usual treatments for patients with non-small-cell lung cancer (NSCLC), such as advanced lung adenocarcinoma, are unspecific and aggressive, and include lung resection, radiotherapy and chemotherapy. Recently, treatment with monoclonal antibodies and biological inhibitors has emerged as an effective alternative, generating effective results with few side effects. In recent years, several clinical trials using monoclonal antibodies presented potential benefits to NSCLC, and 4 of them are already approved for the treatment of NSCLC, such as cetuximab, bevacizumab, nivolumab and pembrolizumab. Also, biological inhibitors are attractive tolls for biological applications. Among the approved inhibitors are crizotinib, erlotinib, afatinib and gefitinib, and side effects are usually mild to intense. Nevertheless, biological molecule treatments are under development, and several new monoclonal antibodies and biological inhibitors are in trial to treat NSCLC. Also under trial study are as follows: anti-epidermal growth factor receptor (EGFR) antibodies (nimotuzumab and ficlatuzumab), anti-IGF 1 receptor (IGF-1R) monoclonal antibody (figitumumab), anti-NR-LU-10 monoclonal antibody (nofetumomab) as well as antibodies directly affecting the cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) molecule (ipilimumab and tremelimumab), to receptor activator of nuclear factor-kappa B ligand (RANKL) (denosumab) or to polymerase enzyme (veliparib and olaparib). Among new inhibitors under investigation are poly-ADP ribose polymerase (PARP) inhibitors (veliparib and olaparib) and phosphatidylinositol 3-kinase (PI3K) inhibitor (buparlisib). However, the success of immunotherapies still requires extensive research and additional controlled trials to evaluate the long-term benefits and side effects.
Collapse
Affiliation(s)
- Ana P S Silva
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil
| | - Priscila V Coelho
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil
| | - Maristella Anazetti
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil.,b Department of Health Science , Faculty DeVry Metrocamp , Campinas , SP , Brazil
| | - Patricia U Simioni
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil.,c Department of Genetics , Evolution and Bioagents, Institute of Biology, University of Campinas (UNICAMP) , Campinas , SP , Brazil.,d Department of Biochemistry and Microbiology , Institute of Biosciences, Universidade Estadual Paulista, UNESP , Rio Claro , SP , Brazil
| |
Collapse
|
65
|
Longo V, Brunetti O, Gnoni A, Cascinu S, Gasparini G, Lorusso V, Ribatti D, Silvestris N. Angiogenesis in pancreatic ductal adenocarcinoma: A controversial issue. Oncotarget 2016; 7:58649-58658. [PMID: 27462915 PMCID: PMC5295459 DOI: 10.18632/oncotarget.10765] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/13/2016] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) occurs in the majority of cases with early loco-regional spread and distant metastases at diagnosis, leading to dismal prognosis with a 5-year overall survival rate moderately over than 5%. This malignancy is largely resistant to chemotherapy and radiation, but the reasons of the refractoriness to the therapies is still unknown. Evidence is accumulating to indicate that the PDAC microenvironment and vascularity strongly contribute to the clinical features of this disease. In particular, PDAC is characterized by excessive dense extracellular matrix deposition associated to vasculature collapse and hypoxia with low drug delivery, explaining at least partly the low efficacy of antiangiogenic drugs in this cancer. Strategies aimed to modulate tumor stroma favoring vasculature perfusion and chemotherapeutics delivery are under investigation.
Collapse
Affiliation(s)
- Vito Longo
- Department of Medical Oncology, Hospital of Taranto, Taranto, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit, Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Antonio Gnoni
- Department of Medical Oncology, Hospital "Vito Fazi" of Lecce, Lecce, Italy
| | | | | | - Vito Lorusso
- Medical Oncology Unit, Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.,National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Cancer Institute "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
66
|
Hamada T, Nakai Y, Isayama H, Yasunaga H, Matsui H, Takahara N, Mizuno S, Kogure H, Matsubara S, Yamamoto N, Tada M, Koike K. Progression-free survival as a surrogate for overall survival in first-line chemotherapy for advanced pancreatic cancer. Eur J Cancer 2016; 65:11-20. [PMID: 27451020 DOI: 10.1016/j.ejca.2016.05.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/24/2016] [Accepted: 05/15/2016] [Indexed: 02/09/2023]
|
67
|
Abstract
Pancreatic cancer is a highly lethal disease, for which mortality closely parallels incidence. Most patients with pancreatic cancer remain asymptomatic until the disease reaches an advanced stage. There is no standard programme for screening patients at high risk of pancreatic cancer (eg, those with a family history of pancreatic cancer and chronic pancreatitis). Most pancreatic cancers arise from microscopic non-invasive epithelial proliferations within the pancreatic ducts, referred to as pancreatic intraepithelial neoplasias. There are four major driver genes for pancreatic cancer: KRAS, CDKN2A, TP53, and SMAD4. KRAS mutation and alterations in CDKN2A are early events in pancreatic tumorigenesis. Endoscopic ultrasonography and endoscopic ultrasonography-guided fine-needle aspiration offer high diagnostic ability for pancreatic cancer. Surgical resection is regarded as the only potentially curative treatment, and adjuvant chemotherapy with gemcitabine or S-1, an oral fluoropyrimidine derivative, is given after surgery. FOLFIRINOX (fluorouracil, folinic acid [leucovorin], irinotecan, and oxaliplatin) and gemcitabine plus nanoparticle albumin-bound paclitaxel (nab-paclitaxel) are the treatments of choice for patients who are not surgical candidates but have good performance status.
Collapse
MESH Headings
- Albumins/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- CA-19-9 Antigen/metabolism
- Camptothecin/administration & dosage
- Camptothecin/analogs & derivatives
- Carcinoembryonic Antigen/metabolism
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/therapy
- Deoxycytidine/administration & dosage
- Deoxycytidine/analogs & derivatives
- Endoscopic Ultrasound-Guided Fine Needle Aspiration
- Endosonography
- Fluorouracil/administration & dosage
- Genes, p16
- Humans
- Irinotecan
- Leucovorin/administration & dosage
- Neoplasms, Cystic, Mucinous, and Serous/diagnosis
- Neoplasms, Cystic, Mucinous, and Serous/genetics
- Neoplasms, Cystic, Mucinous, and Serous/therapy
- Organoplatinum Compounds/administration & dosage
- Oxaliplatin
- Paclitaxel/administration & dosage
- Pancreatectomy
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/therapy
- Proto-Oncogene Proteins p21(ras)/genetics
- Smad4 Protein/genetics
- Tumor Suppressor Protein p53/genetics
- Gemcitabine
Collapse
Affiliation(s)
- Terumi Kamisawa
- Department of Internal Medicine, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan.
| | - Laura D Wood
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, USA
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo, Japan
| | - Kyoichi Takaori
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
68
|
Blanco‐Míguez A, Gutiérrez‐Jácome A, Pérez‐Pérez M, Pérez‐Rodríguez G, Catalán‐García S, Fdez‐Riverola F, Lourenço A, Sánchez B. From amino acid sequence to bioactivity: The biomedical potential of antitumor peptides. Protein Sci 2016; 25:1084-95. [PMID: 27010507 PMCID: PMC4941772 DOI: 10.1002/pro.2927] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/18/2016] [Accepted: 03/22/2016] [Indexed: 12/25/2022]
Abstract
Chemoprevention is the use of natural and/or synthetic substances to block, reverse, or retard the process of carcinogenesis. In this field, the use of antitumor peptides is of interest as, (i) these molecules are small in size, (ii) they show good cell diffusion and permeability, (iii) they affect one or more specific molecular pathways involved in carcinogenesis, and (iv) they are not usually genotoxic. We have checked the Web of Science Database (23/11/2015) in order to collect papers reporting on bioactive peptide (1691 registers), which was further filtered searching terms such as "antiproliferative," "antitumoral," or "apoptosis" among others. Works reporting the amino acid sequence of an antiproliferative peptide were kept (60 registers), and this was complemented with the peptides included in CancerPPD, an extensive resource for antiproliferative peptides and proteins. Peptides were grouped according to one of the following mechanism of action: inhibition of cell migration, inhibition of tumor angiogenesis, antioxidative mechanisms, inhibition of gene transcription/cell proliferation, induction of apoptosis, disorganization of tubulin structure, cytotoxicity, or unknown mechanisms. The main mechanisms of action of those antiproliferative peptides with known amino acid sequences are presented and finally, their potential clinical usefulness and future challenges on their application is discussed.
Collapse
Affiliation(s)
- Aitor Blanco‐Míguez
- ESEI ‐ Escuela Superior De Ingeniería Informática, Edificio Politécnico, Campus Universitario as Lagoas S/N, Universidad De VigoOurense32004Spain
| | - Alberto Gutiérrez‐Jácome
- ESEI ‐ Escuela Superior De Ingeniería Informática, Edificio Politécnico, Campus Universitario as Lagoas S/N, Universidad De VigoOurense32004Spain
| | - Martín Pérez‐Pérez
- ESEI ‐ Escuela Superior De Ingeniería Informática, Edificio Politécnico, Campus Universitario as Lagoas S/N, Universidad De VigoOurense32004Spain
| | - Gael Pérez‐Rodríguez
- ESEI ‐ Escuela Superior De Ingeniería Informática, Edificio Politécnico, Campus Universitario as Lagoas S/N, Universidad De VigoOurense32004Spain
| | - Sandra Catalán‐García
- Asturias, INDRA Software LabsC/Jimena Fernández De La Vega, 140 P. Científico Tecnológico, EdGijón33203Spain
| | - Florentino Fdez‐Riverola
- ESEI ‐ Escuela Superior De Ingeniería Informática, Edificio Politécnico, Campus Universitario as Lagoas S/N, Universidad De VigoOurense32004Spain
| | - Anália Lourenço
- ESEI ‐ Escuela Superior De Ingeniería Informática, Edificio Politécnico, Campus Universitario as Lagoas S/N, Universidad De VigoOurense32004Spain
- Centre of Biological Engineering, University of MinhoCampus De GualtarBraga4710‐057Portugal
| | - Borja Sánchez
- Department of Microbiology and Biochemistry of Dairy ProductsInstituto De Productos Lácteos De Asturias (IPLA), Consejo Superior De Investigaciones Científicas (CSIC)VillaviciosaAsturiasSpain
| |
Collapse
|
69
|
Systematic review and meta-analysis on targeted therapy in advanced pancreatic cancer. Pancreatology 2016; 16:249-58. [PMID: 26852170 DOI: 10.1016/j.pan.2016.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/14/2015] [Accepted: 01/08/2016] [Indexed: 12/11/2022]
Abstract
AIM A systematic review and meta-analysis from literature has been performed to assess the impact of targeted therapy in advanced pancreatic cancer. METHODS By searching different literature databases and major cancer meetings proceedings, data from all randomized clinical trials designed to investigate molecular targeted agents in the treatment of advanced pancreatic cancer were collected. The time-frame between January 2007 and March 2015 was selected. Data on predefined end-points, including overall survival, progression-free survival in terms of Hazard Ratio and response-rate were extracted and analyzed by a random effects model. Pooled data analysis was performed according to the DerSimonian and Laird test. The occurrence of publication bias was investigated through Begg's test by visual inspection of funnel plots. RESULTS Twenty-seven randomized clinical trials for a total of 8205 patients were selected and included in the final analysis. A significant benefit was demonstrated for anti-EGFR agents on overall survival (HR = 0.880; 95% confidence interval (CI) 0.797-0.972; p = 0.011). In the pooled analysis no benefit on overall survival (OS: pooled HR = 0.957; 95%CI 0.900-1.017; p = 0.153), or progression-free survival (PFS: pooled HR = 0.908; 95%CI 0.817-1.010; p = 0.075) for targeted-based therapies as compared to conventional treatments could be demonstrated. No advantage was reported in response-rate (OR for RR = 1.210; 95%CI 0.990-1.478; p = 0.063). Begg's funnel plot showed no evidence of publication bias. CONCLUSION The use of molecular targeted agents does not translate into clinical benefit. Therefore, our work highlights the need to identify predictive factors for patient selection and rationally designed clinical trials.
Collapse
|
70
|
Overview of pre-clinical and clinical studies targeting angiogenesis in pancreatic ductal adenocarcinoma. Cancer Lett 2015; 381:201-10. [PMID: 26723874 DOI: 10.1016/j.canlet.2015.11.047] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/13/2015] [Accepted: 11/30/2015] [Indexed: 12/18/2022]
Abstract
The importance of angiogenesis in pancreatic ductal adenocarcinoma (PDAC) and its therapeutic potential have been explored in both pre-clinical and clinical studies. Human PDACs overexpress a number of angiogenic factors and their cognate high-affinity receptors, and anti-angiogenic agents reduce tumor volume, metastasis, and microvessel density (MVD), and improve survival in subcutaneous and orthotopic pre-clinical models. Nonetheless, clinical trials using anti-angiogenic therapy have been overwhelmingly unsuccessful. This review will focus on these pre-clinical and clinical studies, the potential reasons for failure in the clinical setting, and ways these shortcomings could be addressed in future investigations of angiogenic mechanisms in PDAC.
Collapse
|