1
|
Cao Z, Deng K, Jiang J, Tian K, Wang B. Combined treatment of small cell lung cancer using radiotherapy and immunotherapy: Challenges and updates. Biomed Pharmacother 2025; 182:117727. [PMID: 39675137 DOI: 10.1016/j.biopha.2024.117727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/17/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024] Open
Abstract
Currently, chemotherapy remains the standard first- and second-line treatment for small cell lung cancer (SCLC). Research concerning immunotherapy has brought about a remarkable development in the treatment pattern of SCLC. Atirizumab, duvalizumab, atezolizumab, and serplulimab can significantly improve the clinical outcomes of SCLC. Given the rapidly evolving concept that combining immunotherapy with radiotherapy can increase therapeutic effectiveness, clinicians are devoted to further improving local tumor control by integrating immunotherapy with radiotherapy. This paper reviews the research progress in this field to date and explores ways to further enhance the efficacy of this combination therapy. We first discussed that immunotherapy combined with radiotherapy can improve the abscopal effect, progression-free survival, and overall survival rates of SCLC patients. Then, the biomarkers related to the radiation immune microenvironment, such as programmed death ligand-1 (PD-L1), tumor mutational burden (TMB), and the immune function of patients were discussed. Next, we explored the occurrence and underlying mechanisms of immune resistance during radiotherapy implementation. Finally, we clarified that the emerging trend of low-dose radiotherapy help overcome the inhibitory signals that limit T-cell infiltration in the tumor matrix. In summary, considering the rapid development of this field, these combined therapy strategies may have unlimited potential to further improve the efficacy of radiotherapy combined with immunotherapy for patients.
Collapse
Affiliation(s)
- Zhumin Cao
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing 400054, China.
| | - Kai Deng
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing 400061, China.
| | - Jinxiu Jiang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing 400061, China.
| | - Ke Tian
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing 400054, China.
| | - Bin Wang
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing 400054, China.
| |
Collapse
|
2
|
Lyng FM, Azzam EI. Abscopal Effects, Clastogenic Effects and Bystander Effects: 70 Years of Non-Targeted Effects of Radiation. Radiat Res 2024; 202:355-367. [PMID: 38986531 DOI: 10.1667/rade-24-00040.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/29/2024] [Indexed: 07/12/2024]
Abstract
In vitro and in vivo observations accumulated over several decades have firmly shown that the biological effects of ionizing radiation can spread from irradiated cells/tissues to non-targeted cells/tissues. Redox-modulated intercellular communication mechanisms that include a role for secreted factors and gap junctions, can mediate these non-targeted effects. Clearly, the expression of such effects and their transmission to progeny cells has implications for issues related to radiation protection. Their elucidation is also relevant towards enhancing the efficacy of cancer radiotherapy and reducing its impact on the development of normal tissue toxicities. In addition, the study of non-targeted effects is pertinent to our basic understanding of intercellular communications under conditions of oxidative stress. This review will trace the history of non-targeted effects of radiation starting with early reports of abscopal effects which described radiation induced effects in tissues distant from the site of radiation exposure. A related effect involved the production of clastogenic factors in plasma following irradiation which can induce chromosome damage in unirradiated cells. Despite these early reports suggesting non-targeted effects of radiation, the classical paradigm that a direct deposition of energy in the nucleus was required still dominated. This paradigm was challenged by papers describing radiation induced bystander effects. This review will cover mechanisms of radiation-induced bystander effects and the potential impacts on radiation protection and radiation therapy.
Collapse
Affiliation(s)
- Fiona M Lyng
- Radiation and Environmental Science Centre, FOCAS Research Institute
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Dublin, Ireland
| | - Edouard I Azzam
- Department of Radiology, Rutgers New Jersey Medical School Cancer Center, Newark, New Jersey
| |
Collapse
|
3
|
Li H, Zhao Y, Ma T, Shao H, Wang T, Jin S, Liu Z. Radiotherapy for extensive-stage small-cell lung cancer in the immunotherapy era. Front Immunol 2023; 14:1132482. [PMID: 37701437 PMCID: PMC10493776 DOI: 10.3389/fimmu.2023.1132482] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 08/01/2023] [Indexed: 09/14/2023] Open
Abstract
Currently, chemoimmunotherapy is the first-line treatment for extensive-stage small-cell lung cancer (ES-SCLC). However, only 0.8%-2.5% of the patients presented complete response after chemoimmunotherapy. Considering that ES-SCLC is highly sensitive to radiotherapy, the addition of radiotherapy after first-line treatment for ES-SCLC could further improve local control, which may be beneficial for patients' survival. Prior studies have shown that consolidative thoracic radiotherapy (cTRT) can decrease disease progression and improve overall survival in patients with ES-SCLC who respond well to chemotherapy. However, the efficacy and safety of cTRT in the immunotherapy era remain unclear owing to a lack of prospective studies. Prophylactic cranial irradiation (PCI) has been shown to decrease brain metastasis (BM) and prolong survival in patients with limited-stage SCLC in previous reports. However, according to current guidelines, PCI is not commonly recommended for ES-SCLC. Immunotherapy has the potential to reduce the incidence of BM. Whether PCI can be replaced with regular magnetic resonance imaging surveillance for ES-SCLC in the era of immunotherapy remains controversial. Whole brain radiation therapy (WBRT) is the standard treatment for BM in SCLC patients. Stereotactic radiosurgery (SRS) has shown promise in the treatment of limited BM. Considering the potential of immunotherapy to decrease BM, it is controversial whether SRS can replace WBRT for limited BM in the immunotherapy era. Additionally, with the addition of immunotherapy, the role of palliative radiotherapy may be weakened in patients with asymptomatic metastatic lesions. However, it is still indispensable and urgent for patients with obvious symptoms of metastatic disease, such as spinal cord compression, superior vena cava syndrome, lobar obstruction, and weight-bearing metastases, which may critically damage the quality of life and prognosis. To improve the outcome of ES-SCLC, we discuss the feasibility of radiotherapy, including cTRT, PCI, WBRT/SRS, and palliative radiotherapy with immunotherapy based on existing evidence, which may offer specific prospects for further randomized trials and clinical applications.
Collapse
Affiliation(s)
- Huanhuan Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Jilin University, Changchun, China
| | - Yangzhi Zhao
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Tiangang Ma
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Jilin University, Changchun, China
| | - Hao Shao
- Department of Radiation Oncology, The Second Affiliated Hospital of Jilin University, Changchun, China
| | - Tiejun Wang
- Department of Radiation Oncology, The Second Affiliated Hospital of Jilin University, Changchun, China
| | - Shunzi Jin
- NHC Key Laboratory of Radiobiology, Jilin University, Changchun, China
| | - Zhongshan Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
He J, Hu Q. Progress in the clinical application of immune checkpoint inhibitors in small cell lung cancer. Front Immunol 2023; 14:1126582. [PMID: 37063927 PMCID: PMC10090448 DOI: 10.3389/fimmu.2023.1126582] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Small cell lung cancer (SCLC) is a refractory cancer with poor prognosis due to its aggressive malignancy and high rates of metastasis, recurrence and drug resistance. These characteristics have also greatly impeded the identification of new treatment methods and drugs. The traditional model of SCLC treatment that has been reliant on platinum combined with etoposide for decades has been superseded by the emergence of immune checkpoint inhibitors (ICIs), which have shown significant therapeutic effects and broad application prospects as a monotherapy. This has led to the evaluation of ICIs with different mechanisms of action and their use in combination with radiotherapy or a variety of molecular targeted drugs to achieve synergy, complementary advantages, and reduce adverse reactions. Here, we review the progress in the use of ICIs as a monotherapy or in combination therapy for SCLC and consider the current limitations of these approaches as well as prospects for future developments.
Collapse
|
5
|
Guaitoli G, Neri G, Cabitza E, Natalizio S, Mastrodomenico L, Talerico S, Trudu L, Lauro C, Chiavelli C, Baschieri MC, Bruni A, Dominici M, Bertolini F. Dissecting Immunotherapy Strategies for Small Cell Lung Cancer: Antibodies, Ionizing Radiation and CAR-T. Int J Mol Sci 2022; 23:12728. [PMID: 36361523 PMCID: PMC9656696 DOI: 10.3390/ijms232112728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 12/10/2023] Open
Abstract
Small cell lung cancer (SCLC) is a highly aggressive malignancy that accounts for about 14% of all lung cancers. Platinum-based chemotherapy has been the only available treatment for a long time, until the introduction of immune checkpoint inhibitors (ICIs) recently changed first-line standard of care and shed light on the pivotal role of the immune system. Despite improved survival in a subset of patients, a lot of them still do not benefit from first-line chemo-immunotherapy, and several studies are investigating whether different combination strategies (with both systemic and local treatments, such as radiotherapy) may improve patient outcomes. Moreover, research of biomarkers that may be used to predict patients' outcomes is ongoing. In addition to ICIs, immunotherapy offers other different strategies, including naked monoclonal antibodies targeting tumor associated antigens, conjugated antibody, bispecific antibodies and cellular therapies. In this review, we summarize the main evidence available about the use of immunotherapy in SCLC, the rationale behind combination strategies and the studies that are currently ongoing in this setting, in order to give the reader a clear and complete view of this rapidly expanding topic.
Collapse
Affiliation(s)
- Giorgia Guaitoli
- PhD Program Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giovanni Neri
- PhD Program Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Eleonora Cabitza
- Division of Oncology, Department of Oncology and Hematology, Modena University Hospital, 41124 Modena, Italy
| | - Salvatore Natalizio
- Division of Oncology, Department of Oncology and Hematology, Modena University Hospital, 41124 Modena, Italy
| | - Luciana Mastrodomenico
- Division of Oncology, Department of Oncology and Hematology, Modena University Hospital, 41124 Modena, Italy
| | - Sabrina Talerico
- Division of Oncology, Department of Oncology and Hematology, Modena University Hospital, 41124 Modena, Italy
| | - Lucia Trudu
- Division of Oncology, Department of Oncology and Hematology, Modena University Hospital, 41124 Modena, Italy
| | - Chiara Lauro
- Radiotherapy Unit, Department of Oncology and Hematology, Modena University Hospital, 41124 Modena, Italy
| | - Chiara Chiavelli
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Maria Cristina Baschieri
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Alessio Bruni
- Radiotherapy Unit, Department of Oncology and Hematology, Modena University Hospital, 41124 Modena, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
- Division of Oncology, Department of Oncology and Hematology, Modena University Hospital, 41124 Modena, Italy
| | - Federica Bertolini
- Division of Oncology, Department of Oncology and Hematology, Modena University Hospital, 41124 Modena, Italy
| |
Collapse
|
6
|
Russell J, Fanchon L, Alwaseem H, Molina H, O’Donoghue I, Bahr A, de Stanchina E, Pillarsetty N, Humm JL. Analysis of capecitabine metabolites in conjunction with digital autoradiography in a murine model of pancreatic cancer suggests extensive drug penetration through the tumor. Pharmacol Res Perspect 2022; 10:e00898. [PMID: 35257504 PMCID: PMC8902142 DOI: 10.1002/prp2.898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 12/01/2022] Open
Abstract
Previously published digital autoradiography of 3 H-labeled capecitabine reveals a near-uniform distribution of activity throughout a murine pancreatic model. This is in contrast both to 14 C-labeled gemcitabine, and established expectations, as the dense stroma of pancreatic cancer is understood to inhibit drug penetration. Capecitabine is a pro-drug for 5 FU. The positioning of the radiolabel on capecitabine leaves open the possibility that much of the autoradiographic signal is generated by nontoxic compounds. Studies were performed on tumors derived via organoid culture from a murine KPC tumor. As before, we performed autoradiography comparing 3 H capecitabine to the gemcitabine analog 18 F-FAC. The metabolism of capecitabine in this model was studied through LC-MS of tumor tissue. The autoradiographs confirmed that the 3 H label from capecitabine was much more uniformly distributed through the tumor than the 18 F from the gemcitabine analog. LC-MS revealed that approximately 75% of the molar mass of capecitabine had been converted into 5 FU or pre-5 FU compounds. The remainder had been converted into nontoxic species. Therapeutically relevant capecitabine metabolites achieve a relatively even distribution in this pancreatic cancer model, in contrast to the gemcitabine analog 18 F-FAC. In a human xenograft model, (BxPC3), the 3 H label from capecitabine was also uniformly spread across the tumor autoradiographs. However, at 2 h post-administration the metabolism of capecitabine had proceeded further and the bulk of the agent was in the form of nontoxic species.
Collapse
Affiliation(s)
- James Russell
- Department of Medical PhysicsMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Louise Fanchon
- Department of Medical PhysicsMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Hanan Alwaseem
- The Proteomics Resource CenterThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Henrik Molina
- The Proteomics Resource CenterThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Isabella O’Donoghue
- Department of Medical PhysicsMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Amber Bahr
- Anti‐Tumor Assessment Core FacilityMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Elisa de Stanchina
- Anti‐Tumor Assessment Core FacilityMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | | | - John L. Humm
- Department of Medical PhysicsMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| |
Collapse
|
7
|
Arru C, De Miglio MR, Cossu A, Muroni MR, Carru C, Zinellu A, Paliogiannis P. Durvalumab Plus Tremelimumab in Solid Tumors: A Systematic Review. Adv Ther 2021; 38:3674-3693. [PMID: 34105088 PMCID: PMC8279985 DOI: 10.1007/s12325-021-01796-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/15/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Cancer immunotherapy represents one of the most important innovations in modern medicine. Durvalumab is an anti-programmed cell death ligand 1 (PDL-1) agent which is currently under investigation in several studies in combination with the anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4) drug tremelimumab. The aim of this review was to systematically identify and revise the current scientific literature investigating the combination of these two drugs in solid tumors. METHODS A digital search on the Medline (PubMed interface) and Scopus databases for articles published from inception to 26 February 2021 was performed. The terms used for the search were durvalumab AND tremelimumab. Trials reported in English involving adult patients with solid cancers treated with the combination durvalumab plus tremelimumab were retrieved; the references of the articles were cross-checked to identify missing papers. RESULTS The electronic search produced 267 results; after exclusion of duplicates, irrelevant articles, reviews, and papers not in English or missing data, 19 articles were included for revision. The total number of patients treated with the combination of durvalumab and tremelimumab in the studies retrieved was 2052. CONCLUSION The combination of durvalumab plus tremelimumab showed some oncological advantages in comparison with traditional chemotherapies in some subsets of tumors, but generally has not shown consistent advantages in comparison with the employment of durvalumab monotherapy. A number of the studies examined had intrinsic methodological limitations; therefore, future well-designed studies involving larger cohorts are warranted.
Collapse
Affiliation(s)
- Caterina Arru
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Maria Rosaria De Miglio
- Department of Medical, Clinical and Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy.
| | - Antonio Cossu
- Department of Medical, Clinical and Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Maria Rosaria Muroni
- Department of Medical, Clinical and Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
- Laboratory Quality Control Unit, University Hospital Sassari (AOU-SS), 07100, Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Panagiotis Paliogiannis
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
- Laboratory Quality Control Unit, University Hospital Sassari (AOU-SS), 07100, Sassari, Italy
| |
Collapse
|
8
|
Mallya K, Gautam SK, Aithal A, Batra SK, Jain M. Modeling pancreatic cancer in mice for experimental therapeutics. Biochim Biophys Acta Rev Cancer 2021; 1876:188554. [PMID: 33945847 DOI: 10.1016/j.bbcan.2021.188554] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy that is characterized by early metastasis, low resectability, high recurrence, and therapy resistance. The experimental mouse models have played a central role in understanding the pathobiology of PDAC and in the preclinical evaluation of various therapeutic modalities. Different mouse models with targetable pathological hallmarks have been developed and employed to address the unique challenges associated with PDAC progression, metastasis, and stromal heterogeneity. Over the years, mouse models have evolved from simple cell line-based heterotopic and orthotopic xenografts in immunocompromised mice to more complex and realistic genetically engineered mouse models (GEMMs) involving multi-gene manipulations. The GEMMs, mostly driven by KRAS mutation(s), have been widely accepted for therapeutic optimization due to their high penetrance and ability to recapitulate the histological, molecular, and pathological hallmarks of human PDAC, including comparable precursor lesions, extensive metastasis, desmoplasia, perineural invasion, and immunosuppressive tumor microenvironment. Advanced GEMMs modified to express fluorescent proteins have allowed cell lineage tracing to provide novel insights and a new understanding about the origin and contribution of various cell types in PDAC pathobiology. The syngeneic mouse models, GEMMs, and target-specific transgenic mice have been extensively used to evaluate immunotherapies and study therapy-induced immune modulation in PDAC yielding meaningful results to guide various clinical trials. The emerging mouse models for parabiosis, hepatic metastasis, cachexia, and image-guided implantation, are increasingly appreciated for their high translational significance. In this article, we describe the contribution of various experimental mouse models to the current understanding of PDAC pathobiology and their utility in evaluating and optimizing therapeutic modalities for this lethal malignancy.
Collapse
Affiliation(s)
- Kavita Mallya
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Shailendra K Gautam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
9
|
Jiang Y, Martin J, Alkadhimi M, Shigemori K, Kinchesh P, Gilchrist S, Kersemans V, Smart S, Thompson JM, Hill MA, O'Connor MJ, Davies BR, Ryan AJ. Olaparib increases the therapeutic index of hemithoracic irradiation compared with hemithoracic irradiation alone in a mouse lung cancer model. Br J Cancer 2021; 124:1809-1819. [PMID: 33742147 PMCID: PMC8144220 DOI: 10.1038/s41416-021-01296-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 12/27/2020] [Accepted: 01/27/2021] [Indexed: 12/15/2022] Open
Abstract
Background The radiosensitising effect of the poly(ADP-ribose) polymerase inhibitor olaparib on tumours has been reported. However, its effect on normal tissues in combination with radiation has not been well studied. Herein, we investigated the therapeutic index of olaparib combined with hemithoracic radiation in a urethane-induced mouse lung cancer model. Methods To assess tolerability, A/J mice were treated with olaparib plus whole thorax radiation (13 Gy), body weight changes were monitored and normal tissue effects were assessed by histology. In anti-tumour (intervention) studies, A/J mice were injected with urethane to induce lung tumours, and were then treated with olaparib alone, left thorax radiation alone or the combination of olaparib plus left thorax radiation at 8 weeks (early intervention) or 18 weeks (late intervention) after urethane injection. Anti-tumour efficacy and normal tissue effects were assessed by visual inspection, magnetic resonance imaging and histology. Results Enhanced body weight loss and oesophageal toxicity were observed when olaparib was combined with whole thorax but not hemithorax radiation. In both the early and late intervention studies, olaparib increased the anti-tumour effects of hemithoracic irradiation without increasing lung toxicity. Conclusions The addition of olaparib increased the therapeutic index of hemithoracic radiation in a mouse model of lung cancer.
Collapse
Affiliation(s)
- Yanyan Jiang
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Jennifer Martin
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Maryam Alkadhimi
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Kay Shigemori
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Paul Kinchesh
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Stuart Gilchrist
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Veerle Kersemans
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Sean Smart
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - James M Thompson
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Mark A Hill
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | | | | | - Anderson J Ryan
- CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
10
|
Pakkala S, Higgins K, Chen Z, Sica G, Steuer C, Zhang C, Zhang G, Wang S, Hossain MS, Nazha B, Beardslee T, Khuri FR, Curran W, Lonial S, Waller EK, Ramalingam S, Owonikoko TK. Durvalumab and tremelimumab with or without stereotactic body radiation therapy in relapsed small cell lung cancer: a randomized phase II study. J Immunother Cancer 2020; 8:jitc-2020-001302. [PMID: 33428583 PMCID: PMC7754662 DOI: 10.1136/jitc-2020-001302] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2020] [Indexed: 12/18/2022] Open
Abstract
Background Immune checkpoint blockade (ICB) targeting programmed cell death protein 1 and cytotoxic T lymphocyte-associated protein 4 has achieved modest clinical activity as salvage therapy in relapsed small cell lung cancer (SCLC). We conducted this signal-finding study to assess the efficacy of ICB with or without radiation in relapsed SCLC. Methods Patients with relapsed SCLC and ≤2 previous lines of therapy were randomized to (1) arm A: durvalumab (D) 1500 mg/tremelimumab (T) 75 mg (intravenously every 4 weeks without stereotactic body radiation therapy (SBRT)) or (2) arm B: immune-sensitizing SBRT to one selected tumor site (9 Gy × 3 fractions) followed by D/T. Treatment continued until progression or a maximum of 12 months. The co-primary endpoints of the study were overall response rate (ORR) and progression-free survival (PFS). We evaluated circulating lymphocyte repertoire in serial peripheral blood samples and tumor infiltrating lymphocytes (TILs) from on-treatment biopsies as pharmacodynamic markers. Results Eighteen patients were randomized to arms A and B (n=9 each): median age 70 years; 41.2% women. The median PFS and ORR were 2.1 months and 0% in arm A and 3.3 months and 28.6% in arm B. The median overall survival (OS) was 2.8 months in arm A and 5.7 months in arm B (p=0.3772). Pooled efficacy of D/T±SBRT in 15 Response evaluation criteria in solid tumors (RECIST) evaluable patients across both arms showed the best ORR in terms of partial response in 13.3%, stable disease in 26.6% and progressive disease in 60.0%; the overall median PFS and OS were 2.76 and 3.9 months. The most common adverse events were grade 1 fatigue (66%) and grade 1 elevated amylase (56%) in arm A, and grade 1 fatigue (56%) and pain (44%) in arm B. There was a significant increase in activated CD8(+)ICOS+ T cells (p=0.048) and a reduction in naïve T cells (p=0.0454) in peripheral blood following treatment, along with a significant amount of activated CD8+ICOS+ T cells in TILs from responders. Conclusions The D/T combination with and without SBRT was safe but did not show sufficient efficacy signal in relapsed SCLC. Changes in peripheral blood lymphocyte and TILs were consistent with an immunologic response. Trial registration number NCT02701400.
Collapse
Affiliation(s)
- Suchita Pakkala
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Kristin Higgins
- Department of Radiation Oncology, Emory University, Atlanta, Georgia, USA
| | - Zhengjia Chen
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Gabriel Sica
- Pathology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Conor Steuer
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Chao Zhang
- Biostatistics, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Guojing Zhang
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Shuhua Wang
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Mohammad S Hossain
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Bassel Nazha
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Tyler Beardslee
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Fadlo R Khuri
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Walter Curran
- Department of Radiation Oncology, Emory University, Atlanta, Georgia, USA
| | - Sagar Lonial
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Edmund K Waller
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Suresh Ramalingam
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Taofeek K Owonikoko
- Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| |
Collapse
|
11
|
Immunostimulatory Effects of Radiotherapy for Local and Systemic Control of Melanoma: A Review. Int J Mol Sci 2020; 21:ijms21239324. [PMID: 33297519 PMCID: PMC7730562 DOI: 10.3390/ijms21239324] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
Recently, modern therapies involving immune checkpoint inhibitors, cytokines, and oncolytic virus have been developed. Because of the limited treatment effect of modern therapy alone, the immunostimulatory effect of radiotherapy attracted increasing attention. The combined use of radiotherapy and modern therapy has been examined clinically and non-clinically, and its effectiveness has been confirmed recently. Because melanomas have high immunogenicity, better therapeutic outcomes are desired when using immunotherapy. However, sufficient therapeutic effects have not yet been achieved. Thus far, radiotherapy has been used only for local control of tumors. Although extremely rare, radiotherapy has also been reported for systemic control, i.e., abscopal effect. This is thought to be due to an antitumor immune response. Therefore, we herein summarize past information on not only the mechanism of immune effects on radiotherapy but also biomarkers reported in case reports on abscopal effects. We also reviewed the animal model suitable for evaluating abscopal effects. These results pave the way for further basic research or clinical studies on new treatment methods for melanoma. Currently, palliative radiation is administered to patients with metastatic melanoma for local control. If it is feasible to provide both systemic and local control, the treatment benefit for the patients is very large.
Collapse
|
12
|
Higgins KA, Slotman BJ. What is the role of consolidative thoracic radiotherapy in the era of chemo-immunotherapy for extensive stage small cell lung cancer? J Thorac Dis 2020; 12:6308-6310. [PMID: 33209469 PMCID: PMC7656365 DOI: 10.21037/jtd.2020.03.15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Kristin A Higgins
- Department of Radiation Oncology, Emory University, Atlanta, GA, USA.,Department of Radiation Oncology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ben J Slotman
- Department of Radiation Oncology, Emory University, Atlanta, GA, USA.,Department of Radiation Oncology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Chen YH, Chen YC, Lin CC, Hsieh YP, Hsu CS, Hsieh MC. Synergistic Anticancer Effects of Gemcitabine with Pitavastatin on Pancreatic Cancer Cell Line MIA PaCa-2 in vitro and in vivo. Cancer Manag Res 2020; 12:4645-4665. [PMID: 32606957 PMCID: PMC7306478 DOI: 10.2147/cmar.s247876] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/13/2020] [Indexed: 12/21/2022] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with an overall 5-year survival rate of 9.3%, and this malignancy is expected to become the second leading cause of cancer-related death by 2030. Gemcitabine resistance develops within weeks of PDAC patient’s chemotherapeutic initiation. Statins, including pitavastatin, have been indicated to have anticancer effects in numerous human cancer cell lines. Thus, in this study, we hypothesized that a combination of gemcitabine and pitavastatin may have a greater anticancer effect than gemcitabine alone on the human pancreatic carcinoma cell line MIA PaCa-2. Methods The anticancer effects of gemcitabine with pitavastatin were evaluated using human MIA PaCa-2 cell line in vitro and in vivo Balb/c murine xenograft tumor model. Cell viability was assessed with CCK-8, and cell migration was stained by crystal violet. Cell cycle distribution, apoptosis and mitochondrial membrane potential were examined by flow cytometry. Activation of drug transporters (hENTs, hCNTs), intracellular drug activating (dCK) and inhibition of inactivating enzymes (RRMs) pathways were assessed by Western blotting analysis. Molecular mechanisms and signaling pathways of apoptosis, necrosis and autophagy also were assessed by Western blotting. Results We observed that gemcitabine and pitavastatin synergistically suppressed the proliferation of MIA PaCa-2 cells through causing sub-G1 and S phase cell cycle arrest. Activation of apoptosis/necrosis was confirmed by annexin V/propidium iodide double staining, which showed increasing levels of active caspase 3, cleaved poly(ADP-ribose) polymerase and the RIP1–RIP3–MLKL complex. Moreover, gemcitabine–pitavastatin-mediated S phase arrest downregulated cyclin A2/CDK2 and upregulated p21/p27 in MIA PaCa-2 cells. Furthermore, this combination improved drug cellular metabolism pathway, mitochondria function and activated autophagy as part of the cell death mechanism. In vivo, gemcitabine-pitavastatin effectively inhibited tumor growth in a nude mouse mode of Mia PaCa-2 xenografts without observed adverse effect. Conclusion Combined gemcitabine–pitavastatin may be an effective novel treatment option for pancreatic cancer.
Collapse
Affiliation(s)
- Ya-Hui Chen
- Diabetes Research Laboratory, Department of Research, Changhua Christian Hospital, Changhua, Taiwan.,Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Chun Chen
- Diabetes Research Laboratory, Department of Research, Changhua Christian Hospital, Changhua, Taiwan
| | - Chi-Chen Lin
- Institute of Biomedical Science, National Chung-Hsing University, Taichung, Taiwan.,Department of Health and Nutrition, Asia University, Taichung, Taiwan.,Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Yao-Peng Hsieh
- Division of General Internal Medicine, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan.,School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Sheng Hsu
- Frontier Molecular Medical Research Center in Children, Changhua Christian Children Hospital, Changhua, Taiwan
| | - Ming-Chia Hsieh
- Diabetes Research Laboratory, Department of Research, Changhua Christian Hospital, Changhua, Taiwan.,Intelligent Diabetes Metabolism and Exercise Center, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
14
|
Xie C, Duffy AG, Brar G, Fioravanti S, Mabry-Hrones D, Walker M, Bonilla CM, Wood BJ, Citrin DE, Gil Ramirez EM, Escorcia FE, Redd B, Hernandez JM, Davis JL, Gasmi B, Kleiner D, Steinberg SM, Jones JC, Greten TF. Immune Checkpoint Blockade in Combination with Stereotactic Body Radiotherapy in Patients with Metastatic Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2020; 26:2318-2326. [PMID: 31996388 DOI: 10.1158/1078-0432.ccr-19-3624] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/26/2019] [Accepted: 01/27/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE The effectiveness of immune checkpoint inhibitors (ICI) is limited in pancreatic ductal adenocarcinoma (PDAC). We conducted a phase I study to evaluate the safety of ICI with stereotactic body radiation therapy (SBRT) in patients with metastatic PDAC. PATIENTS AND METHODS Patients enrolled must have received at least one line of prior systemic chemotherapy for metastatic disease. Cohorts A1 and A2 received durvalumab every 2 weeks plus either 8 Gy in one fraction of SBRT on day 1 or 25 Gy in five fractions on day -3 to +1. Cohorts B1 and B2 received durvalumab plus tremelimumab every 4 weeks and either 8 Gy in one fraction of SBRT on day 1 or 25 Gy in five fractions on day -3 to +1. ICIs were continued until unacceptable toxicity or disease progression. The primary objective was the safety and feasibility of treatment. Objective response was assessed in lesions not subjected to SBRT. RESULTS Fifty-nine patients were enrolled and 39 were evaluable for efficacy. No dose-limiting toxicities were seen. The most common adverse event was lymphopenia. Two patients achieved a partial response (one confirmed and the other unconfirmed). The overall response rate was 5.1%. Median PFS and OS was 1.7 months [95% confidence intervals (CI), 0.8-2.0 months] and 3.3 months (95% CI, 1.2-6.6 months) in cohort A1; 2.5 months (95% CI, 0.1-3.7 months) and 9.0 months (95% CI, 0.5-18.4 months) in A2; 0.9 months (95% CI, 0.7-2.1 months) and 2.1 months (95% CI, 1.1-4.3 months) in B1; and 2.3 months (95% CI, 1.9-3.4 months) and 4.2 months (95% CI, 2.9-9.3 months) in B2. CONCLUSIONS The combination of ICI and SBRT has an acceptable safety profile and demonstrates a modest treatment benefit in patients with metastatic PDAC.
Collapse
Affiliation(s)
- Changqing Xie
- Gastrointestinal Malignancy Section, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Austin G Duffy
- Gastrointestinal Malignancy Section, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Gagandeep Brar
- Hematology/Oncology Fellowship Program, National Heart, Lung, and Blood Institute, NCI, NIH, Bethesda, Maryland
| | - Suzanne Fioravanti
- Gastrointestinal Malignancy Section, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Donna Mabry-Hrones
- Gastrointestinal Malignancy Section, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Melissa Walker
- Gastrointestinal Malignancy Section, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Cecilia Monge Bonilla
- Gastrointestinal Malignancy Section, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Bradford J Wood
- Radiology and Imaging Sciences, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Deborah E Citrin
- Radiation Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | | | - Freddy E Escorcia
- Radiation Oncology Branch, Laboratory of Molecular Radiotherapy, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Bernadette Redd
- Radiology and Imaging Sciences, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Jonathan M Hernandez
- Surgical Oncology Program, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Jeremy L Davis
- Surgical Oncology Program, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Billel Gasmi
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - David Kleiner
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Jennifer C Jones
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Center for Cancer Research, NCI, NIH, Bethesda, Maryland. .,NCI CCR Liver Cancer Program, NIH, Bethesda, Maryland
| |
Collapse
|
15
|
Higgins KA, Gorgens S, Sudmeier LJ, Faivre-Finn C. Recent developments in limited stage small cell lung cancer. Transl Lung Cancer Res 2019; 8:S147-S152. [PMID: 31673519 DOI: 10.21037/tlcr.2019.05.13] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Limited stage small cell lung cancer (LS-SCLC) remains a challenging disease, with 5-year overall survival ranging from 30-35% with current standard of care treatment consisting of thoracic radiation to 45 Gy in 30 fractions delivered twice daily, with concurrent platinum/etoposide chemotherapy, followed by prophylactic cranial irradiation (PCI). The randomized, phase III CONVERT study confirmed 45 Gy delivered twice daily to be the optimal radiation fractionation regimen, without significantly increased toxicity when compared to daily radiation to 66 Gy. Immunotherapy is now being studied in addition to chemoradiation, in both the concurrent and consolidative setting. These randomized trials are ongoing. Additionally, the role of PCI compared to MRI surveillance is being evaluated in patients with LS-SCLC in both the North America and Europe. Ideally these ongoing studies will continue to improve outcomes for LS-SCLC.
Collapse
Affiliation(s)
- Kristin A Higgins
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Sophia Gorgens
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Lisa J Sudmeier
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Corinne Faivre-Finn
- Department of Thoracic Oncology, University of Manchester, The Christie NHS Foundation Trust, Withington, Manchester, UK
| |
Collapse
|
16
|
Siddiqui NS, Godara A, Byrne MM, Saif MW. Capecitabine for the treatment of pancreatic cancer. Expert Opin Pharmacother 2019; 20:399-409. [PMID: 30649964 DOI: 10.1080/14656566.2018.1560422] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Capecitabine is an oral prodrug of 5-fluorouracil (5-FU) which is converted to 5FU by a series of reactions catalyzed by different enzymes, the last of the enzymes being thymidine phosphorylase (TP). TP is found to be elevated in tumor cells in comparison to normal cells, which consequently tumor-localizes the production of 5-FU, thereby limiting its systemic toxicity. Today, capecitabine is extensively used for the treatment of many solid malignancies, with a particular focus in breast and gastrointestinal tumors, but also in pancreatic cancer. Areas covered: This review summarizes the pharmacology and the clinical evidence relevant to the use of capecitabine in the treatment of pancreas cancer. The authors provide, furthermore, provide their expert perspectives on its use. Expert opinion: Capecitabine has the advantage over other therapeutics in so much that it has both convenient oral administration and a favorable toxicity profile. Current data has promised the use of capecitabine in all stages of pancreatic cancer. However, predictive markers for outcome and toxicity remain to be validated.
Collapse
Affiliation(s)
- Nauman S Siddiqui
- a Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA
| | - Amandeep Godara
- a Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA
| | - Margaret M Byrne
- a Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA
| | - Muhammad Wasif Saif
- a Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA.,b Department of Medical Oncology , Northwell Health Cancer Institute , New York , NY , USA
| |
Collapse
|
17
|
Immune Checkpoint Inhibitors in Early-Stage and Locally Advanced Non-Small Cell Lung Cancer. Curr Treat Options Oncol 2018; 19:39. [PMID: 29931587 DOI: 10.1007/s11864-018-0556-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OPINION STATEMENT Surgical resection ± chemotherapy ± radiation or stereotactic body radiation therapy (SBRT) are established treatment modalities for resectable stage non-small cell lung cancer (NSCLC), and concurrent chemotherapy with radiation is the therapy of choice for unresectable locally advanced disease. Despite treatment with curative intent, most patients subsequently relapse and develop distant disease. Treatment with checkpoint inhibitors represents a major advancement in the treatment of metastatic NSCLC. Therapy against programed cell death-1/programmed cell death ligand 1 (PD-1/PD-L1) is associated with a significant improvement in overall survival in stage IV disease, and these results have led to a great interest in evaluating these agents in earlier-stage NSCLC. The preliminary data from ongoing trials suggest that the integration of checkpoint blockage into the treatment of early-stage and locally advanced NSCLC is safe, tolerable, and has the potential to improve outcomes without adding substantial toxicity. In the current review, we provide an overview of the emerging data on the role of PD-1/PD-L1 and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) inhibitors in the treatment of early-stage and locally advanced NSCLC, with a focus on ongoing clinical trials and combination strategies.
Collapse
|
18
|
Brix N, Tiefenthaller A, Anders H, Belka C, Lauber K. Abscopal, immunological effects of radiotherapy: Narrowing the gap between clinical and preclinical experiences. Immunol Rev 2018; 280:249-279. [PMID: 29027221 DOI: 10.1111/imr.12573] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Radiotherapy-despite being a local therapy that meanwhile is characterized by an impressively high degree of spatial accuracy-can stimulate systemic phenomena which occasionally lead to regression and rejection of non-irradiated, distant tumor lesions. These abscopal effects of local irradiation have been observed in sporadic clinical case reports since the beginning of the 20th century, and extensive preclinical work has contributed to identify systemic anti-tumor immune responses as the underlying driving forces. Although abscopal tumor regression still remains a rare event in the radiotherapeutic routine, increasing numbers of cases are being reported, particularly since the clinical implementation of immune checkpoint inhibiting agents. Accordingly, interests to systematically exploit the therapeutic potential of radiotherapy-stimulated systemic responses are constantly growing. The present review briefly delineates the history of radiotherapy-induced abscopal effects and the activation of systemic anti-tumor immune responses by local irradiation. We discuss preclinical and clinical reports with specific focus on the corresponding controversies, and we propose issues that should be addressed in the future in order to narrow the gap between preclinical knowledge and clinical experiences.
Collapse
Affiliation(s)
- Nikko Brix
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Anna Tiefenthaller
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Heike Anders
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,German Cancer Consortium Partner Site München, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| |
Collapse
|
19
|
Shafique MR, Robinson LA, Antonia S. Durvalumab: a potential maintenance therapy in surgery-ineligible non-small-cell lung cancer. Cancer Manag Res 2018; 10:931-940. [PMID: 29760563 PMCID: PMC5937504 DOI: 10.2147/cmar.s148009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the most common cancer worldwide and the most common cause of cancer-related death. Non-small-cell lung cancer comprises ~87% of newly diagnosed cases of lung cancer, and nearly one-third of these patients have stage III disease. Despite improvements in the treatment of stage IV lung cancer, particularly with the introduction and dissemination of checkpoint inhibitors, very little progress has been made in the treatment of stage III lung cancer. In this article, we discuss the general staging criteria and treatment options for stage III lung cancer. We review how concurrent radiation and chemotherapy can have immunomodulatory effects, supporting the rationale for incorporating immunotherapy into existing treatment paradigms. Finally, we discuss the results of the PACIFIC trial and implications for the treatment of stage III lung cancer. In the PACIFIC trial, adding durvalumab as a maintenance therapy following the completion of chemoradiotherapy improved progression-free survival in patients with locally advanced unresectable stage III lung cancer. On the strength of these results, durvalumab has been approved by the US Food and Drug Administration for use in this setting, representing the first advance in the treatment of stage III lung cancer in nearly a decade.
Collapse
Affiliation(s)
- Michael R Shafique
- Department of Thoracic Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Lary A Robinson
- Department of Thoracic Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Scott Antonia
- Department of Thoracic Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
20
|
Marconi R, Strolin S, Bossi G, Strigari L. A meta-analysis of the abscopal effect in preclinical models: Is the biologically effective dose a relevant physical trigger? PLoS One 2017; 12:e0171559. [PMID: 28222111 PMCID: PMC5319701 DOI: 10.1371/journal.pone.0171559] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 01/23/2017] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Preclinical in vivo studies using small animals are considered crucial in translational cancer research and clinical implementation of novel treatments. This is of paramount relevance in radiobiology, especially for any technological developments permitted to deliver high doses in single or oligo-fractionated regimens, such as stereotactic ablative radiotherapy (SABR). In this context, clinical success in cancer treatment needs to be guaranteed, sparing normal tissue and preventing the potential spread of disease or local recurrence. In this work we introduce a new dose-response relationship based on relevant publications concerning preclinical models with regard to delivered dose, fractionation schedule and occurrence of biological effects on non-irradiated tissue, abscopal effects. METHODS We reviewed relevant publications on murine models and the abscopal effect in radiation cancer research following PRISMA methodology. In particular, through a log-likelihood method, we evaluated whether the occurrence of abscopal effects may be related to the biologically effective dose (BED). To this aim, studies accomplished with different tumor histotypes were considered in our analysis including breast, colon, lung, fibrosarcoma, pancreas, melanoma and head and neck cancer. For all the tumors, the α / β ratio was assumed to be 10 Gy, as generally adopted for neoplastic cells. RESULTS Our results support the hypothesis that the occurrence rate of abscopal effects in preclinical models increases with BED. In particular, the probability of revealing abscopal effects is 50% when a BED of 60 Gy is generated. CONCLUSION Our study provides evidence that SABR treatments associated with high BEDs could be considered an effective strategy in triggering the abscopal effect, thus shedding light on the promising outcomes revealed in clinical practice.
Collapse
Affiliation(s)
- Raffaella Marconi
- Laboratory of Medical Physics and Expert Systems, Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Strolin
- Laboratory of Medical Physics and Expert Systems, Regina Elena National Cancer Institute, Rome, Italy
| | - Gianluca Bossi
- Laboratory of Medical Physics and Expert Systems, Regina Elena National Cancer Institute, Rome, Italy.,Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy
| | - Lidia Strigari
- Laboratory of Medical Physics and Expert Systems, Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
21
|
Wilks AB, Saif MW. First Case of Foot Drop Associated with Capecitabine in a Patient with Thymidylate Synthase Polymorphism. Cureus 2017; 9:e995. [PMID: 28280649 PMCID: PMC5325748 DOI: 10.7759/cureus.995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Capecitabine, an oral prodrug of 5-FU, has been approved by the FDA for use in patients with breast and colon cancers. In addition, capecitabine is commonly used in patients with other malignancies such as pancreatic, gastroesophageal, and hepatobiliary tract cancers. Though cerebellar toxicity is a rare but well-known side effect of intravenous 5-FU therapy, peripheral neuropathy with capecitabine has only been described in rare cases. In this case report, we describe a 79-year-old patient with locally advanced adenocarcinoma of the pancreas undergoing chemoradiation therapy with capecitabine who developed peripheral sensorimotor neuropathy. To the best of our knowledge, this is the first patient in the literature who was found to have two mutations (2R) of a 28 base-pair tandem repeat in the 5’ promoter enhancer region (5’-TSER) on both alleles (2R/2R) of thymidylate synthetase (TYMS) gene, possibly responsible for the neurotoxicity.
Collapse
Affiliation(s)
- Andrew B Wilks
- Tufts Medical Center, Tufts University School of Medicine
| | - Muhammad W Saif
- Hematology/Oncology, Tufts Medical Center, Tufts University School of Medicine
| |
Collapse
|
22
|
Tsuchikawa T, Takeuchi S, Nakamura T, Shichinohe T, Hirano S. Clinical impact of chemotherapy to improve tumor microenvironment of pancreatic cancer. World J Gastrointest Oncol 2016; 8:786-792. [PMID: 27895816 PMCID: PMC5108980 DOI: 10.4251/wjgo.v8.i11.786] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/19/2016] [Accepted: 09/18/2016] [Indexed: 02/05/2023] Open
Abstract
A perioperative multimodal strategy including combination chemotherapy and radiotherapy, in addition to surgical resection, has been acknowledged to improve patient prognosis. However chemotherapy has not been actively applied as an immunomodulating modality because of concerns about various immunosuppressive effects. It has recently been shown that certain chemotherapeutic agents could modify tumor microenvironment and host immune responses through several underlying mechanisms such as immunogenic cell death, local T-cell infiltration and also the eradication of immune-suppressing regulatory cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells. With the better understanding of the cell components in the tumor microenvironment and the effect of chemotherapy to improve tumor microenvironment, it has been gradually clear that the chemotherapeutic agents is two-edged sword to have both immune promoting and suppressing effects. The cellular components of the tumor microenvironment include infiltrating T lymphocytes, dendritic cells, regulatory T cells, tumor associated macrophages, myeloid derived suppressor cells and cancer associated fibroblasts. Based on the better understanding of tumor microenvironment following chemotherapy, the treatment protocol could be modified as personalized medicine and the prognosis of pancreas cancer would be more improved utilizing multimodal chemotherapy. Here we review the recent advances of chemotherapy to improve tumor microenvironment of pancreatic cancer, introducing the unique feature of tumor microenvironment of pancreatic cancer, interaction between anti-cancer reagents and these constituting cells and future prospects.
Collapse
|
23
|
Cyclooxygenase-2: A Role in Cancer Stem Cell Survival and Repopulation of Cancer Cells during Therapy. Stem Cells Int 2016; 2016:2048731. [PMID: 27882058 PMCID: PMC5108861 DOI: 10.1155/2016/2048731] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/16/2016] [Accepted: 09/26/2016] [Indexed: 12/13/2022] Open
Abstract
Cyclooxygenase-2 (COX-2) is an inducible form of the enzyme that catalyses the synthesis of prostanoids, including prostaglandin E2 (PGE2), a major mediator of inflammation and angiogenesis. COX-2 is overexpressed in cancer cells and is associated with progressive tumour growth, as well as resistance of cancer cells to conventional chemotherapy and radiotherapy. These therapies are often delivered in multiple doses, which are spaced out to allow the recovery of normal tissues between treatments. However, surviving cancer cells also proliferate during treatment intervals, leading to repopulation of the tumour and limiting the effectiveness of the treatment. Tumour cell repopulation is a major cause of treatment failure. The central dogma is that conventional chemotherapy and radiotherapy selects resistant cancer cells that are able to reinitiate tumour growth. However, there is compelling evidence of an active proliferative response, driven by increased COX-2 expression and downstream PGE2 release, which contribute to the repopulation of tumours and poor patient outcome. In this review, we will examine the evidence for a role of COX-2 in cancer stem cell biology and as a mediator of tumour repopulation that can be molecularly targeted to overcome resistance to therapy.
Collapse
|
24
|
Kimbara S, Kondo S. Immune checkpoint and inflammation as therapeutic targets in pancreatic carcinoma. World J Gastroenterol 2016; 22:7440-52. [PMID: 27672267 PMCID: PMC5011660 DOI: 10.3748/wjg.v22.i33.7440] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 06/30/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic adenocarcinoma (PAC) is one of the most deadly malignant neoplasms, and the efficacy of conventional cytotoxic chemotherapy is far from satisfactory. Recent research studies have revealed that immunosuppression and inflammation are associated with oncogenesis, as well as tumor development, invasion, and metastasis in PAC. Thus, immunosuppression-related signaling, especially that involving immune checkpoint and inflammation, has emerged as novel treatment targets for PAC. However, PAC is an immune-resistant tumor, and it is still unclear whether immune checkpoint or anti-inflammation therapies would be an ideal strategy. In this article, we will review immune checkpoint and inflammation as potential targets, as well as clinical trials and the prospects for immunotherapy in PAC.
Collapse
|
25
|
Salehifar E, Hosseinimehr SJ. The use of cyclooxygenase-2 inhibitors for improvement of efficacy of radiotherapy in cancers. Drug Discov Today 2016; 21:654-62. [PMID: 26955911 DOI: 10.1016/j.drudis.2016.02.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 02/02/2016] [Accepted: 02/29/2016] [Indexed: 12/13/2022]
Abstract
Cyclooxygenase-2 (COX-2) is overexpressed in cancer cells and is associated with carcinogenesis and maintenance of progressive tumour growth as well as resistance of cancer cells to ionising radiation (IR). COX-2 inhibitors can attenuate tumour growth and expression of markers of cell proliferation as well as induce apoptosis in tumour cells. These agents can have a synergistic effect with IR in the killing of cancer cells. In this review, we discuss the rational basis and molecular mechanisms regarding the usefulness of COX-2 inhibitors in cancer therapy, and also their potential role in increasing the therapeutic index of chemoradiation by protecting normal cells and sensitising tumour cells to radiotherapy.
Collapse
Affiliation(s)
- Ebrahim Salehifar
- Department of Clinical Pharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
26
|
Wong ET, Lok E, Swanson KD. Clinical benefit in recurrent glioblastoma from adjuvant NovoTTF-100A and TCCC after temozolomide and bevacizumab failure: a preliminary observation. Cancer Med 2015; 4:383-91. [PMID: 25620708 PMCID: PMC4380964 DOI: 10.1002/cam4.421] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 12/24/2014] [Accepted: 01/01/2015] [Indexed: 11/18/2022] Open
Abstract
The NovoTTF-100A is a device that emits alternating electric fields and it is approved for the treatment of recurrent glioblastoma. It works by perturbing tumor cells during mitosis as they enter anaphase leading to aneuploidy, asymmetric chromosome segregation and cell death with evidence of increased immunogenicity. Clinical trial data have shown equivalent efficacy when compared to salvage chemotherapies in recurrent disease. Responders were found to have had a lower dexamethasone usage and a higher rate of prior low-grade histology. We treated a series of patients with NovoTTF-100A and bevacizumab alone (n = 34) or in combination with a regimen consisting of 6-thioguanine, lomustine, capecitabine, and celecoxib (TCCC) (n = 3). Compared to the former cohort, the latter cohort exhibited a trend for prolonged overall survival, median 4.1 (0.3–22.7) months versus 10.3 (7.7–13.6) months respectively (P = 0.0951), with one experiencing an objective response with a 50% reduction in tumor size on magnetic resonance imaging despite possessing a larger tumor size at baseline and more severe neurologic dysfunction than the median for either group. These observations illustrate the possibility of improving survival and achieving a response in patients with end-stage recurrent glioblastoma by biasing the tumor toward anti-tumor immunologic response with a combination of NovoTTF-100A and TCCC, as well as the continuation of bevacizumab in order to limit dexamethasone use due to its global immunosuppressive effect on the patient.
Collapse
Affiliation(s)
- Eric T Wong
- Brain Tumor Center and Neuro-Oncology Unit, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | | |
Collapse
|
27
|
SAIF MUHAMMADWASIF, LEDBETTER LESLIE, KALEY KRISTIN, GARCON MARIECARMEL, RODRIGUEZ TERESA, SYRIGOS KOSTASN. Maintenance therapy with capecitabine in patients with locally advanced unresectable pancreatic adenocarcinoma. Oncol Lett 2014; 8:1302-1306. [PMID: 25120712 PMCID: PMC4114599 DOI: 10.3892/ol.2014.2238] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 02/11/2014] [Indexed: 11/05/2022] Open
Abstract
Therapeutic options for locally advanced pancreatic cancer (LAPC) include concurrent chemoradiation, induction chemotherapy followed by chemoradiation or systemic therapy alone. The original Gastro-Intestinal Study Group and Eastern Cooperative Oncology Group studies defined fluorouracil (5-FU) with concurrent radiation therapy followed by maintenance 5-FU until progression, as the standard therapy for this subset of patients. Although this combined therapy has been demonstrated to increase local control and median survival from 8 to 12 months, almost all patients succumb to the disease secondary to either local or distant recurrence. Our earlier studies provided a strong rationale for the use of capecitabine in combination with concurrent radiation followed by maintenance capecitabine therapy. To report our clinical experience, we retrospectively evaluated our patients who were treated with maintenance capecitabine. We reviewed the medical records of patients with LAPC who received treatment with capecitabine and radiation, followed by a 4-week rest, then capecitabine alone 1,000 mg twice daily (ECOG performance status 2 or age >70 years) or 1,500 mg twice daily for 14 days every 3 weeks until progressive disease. We treated 43 patients between September 2004 and September 2012. The population consisted of 16 females and 25 males, with a median age of 64 years (range, 38-80 years). Patients received maintenance capecitabine for median duration of 9 months (range, 3-18 months). The median overall survival (OS) for these patients was 17 months, with two patients still living and receiving therapy. The 6-month survival rate was 91% (39/43), 1-year survival rate was 72% (31/43) and 2-year OS rate was 26% (11/43). Grade 3 or 4 toxicity was observed rarely: Hand-foot syndrome (HFS) in two patients, diarrhea in one patient and peripheral neuropathy in one patient, and there was no mortality directly related to treatment. Capecitabine maintenance therapy following chemoradiation in LAPC offers an effective, tolerable and convenient alternative to 5-FU. To the best of our knowledge, this is the largest study of its kind which has determined the safety and efficacy of capecitabine maintenance therapy for patients with LAPC.
Collapse
Affiliation(s)
- MUHAMMAD WASIF SAIF
- Section of GI Cancers and Experimental Therapeutics, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | | | | - KOSTAS N. SYRIGOS
- Oncology Unit, Third Department of Medicine, University of Athens, Sotiria General Hospital, Athens 115 27, Greece
| |
Collapse
|
28
|
Sharon E, Polley MY, Bernstein MB, Ahmed M. Immunotherapy and radiation therapy: considerations for successfully combining radiation into the paradigm of immuno-oncology drug development. Radiat Res 2014; 182:252-7. [PMID: 25003314 DOI: 10.1667/rr13707.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
As the immunotherapy of cancer comes of age, adding immunotherapeutic agents to radiation therapy has the potential to improve the outcomes for patients with a wide variety of malignancies. Despite the enormous potential of such combination therapy, laboratory data has been lacking and there is little guidance for pursuing novel treatment strategies. Animal models have significant limitation in combining radiation therapy with immunotherapy and some of the limitations of preclinical models are discussed in this article. In addition to the preclinical challenges, radiation therapy and immunotherapy combinations may have overlapping toxicities, and for both types of therapy, early and late manifestations of toxicity are possible. Given these risks, special attention should be given to the design of the specific Phase I clinical trial that is chosen. In this article, we describe several Phase I design possibilities that may be employed, including the 3 + 3 design (also known as the cohort of 3 design), the continual reassessment method (CRM), and the time-to-event continual reassessment method (TITE-CRM). Efficacy end points for further development of combination therapy must be based on multiple factors, including disease type, stage of disease, the setting of therapy and the goal of therapy. While the designs for future clinical trials will vary, it is clear that these two successful modalities of therapy can and should be combined for the benefit of cancer patients.
Collapse
Affiliation(s)
- Elad Sharon
- a Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | | |
Collapse
|
29
|
Wang H, Zhang L, Shi Y, Javidiparsijani S, Wang G, Li X, Ouyang W, Zhou J, Zhao L, Wang X, Zhang X, Gao F, Liu J, Luo J, Tang J. Abscopal antitumor immune effects of magnet-mediated hyperthermia at a high therapeutic temperature on Walker-256 carcinosarcomas in rats. Oncol Lett 2014; 7:764-770. [PMID: 24527084 PMCID: PMC3919910 DOI: 10.3892/ol.2014.1803] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 12/20/2013] [Indexed: 11/11/2022] Open
Abstract
The abscopal effect has previously been described in various tumors and is associated with radiation therapy and hyperthermia, with possible underlying mechanisms explaining each observed case. In the present study, we aimed to investigate the antitumor effects of magnet-mediated hyperthermia on Walker-256 carcinosarcomas in rats at two different temperature ranges (42–46°C and 50–55°C). We also aimed to identify whether a higher therapeutic temperature of magnetic-mediated hyperthermia improves the abscopal antitumor effects, where localised irradiation of the tumor causes not only the irradiated tumor to shrink, but also tumors located far from the area of irradiation. Following induction of carcinosarcoma in both sides of the body, magnet-mediated hyperthermia was applied to one side only, leaving the other side as a control. The changes in tumor growth were observed. Our results demonstrated that magnet-mediated hyperthermia at a higher temperature inhibited the growth of carcinosarcoma at the site of treatment. Furthermore, the growth of the carcinosarcoma on the untreated side was also inhibited. The expression levels of proliferating cell nuclear antigen were decreased in the hyperthermia group, which was more significant in the higher temperature test group. Flow cytometric analysis showed an increased number of CD4- and CD8-positive T cells, and enzyme-linked immunosorbent assay showed increased levels of interferon-γ and interleukin-2 in the higher temperature group. These results suggested that magnet-mediated hyperthermia at a higher temperature (50–55°C) can improve the abscopal antitumor effects and stimulate a greater endogenous immune response in carcinosarcoma-bearing rats.
Collapse
Affiliation(s)
- Hui Wang
- Department of Radiation Oncology, The Affiliated Hunan Provincial Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China ; Department of Radiation Oncology, The Affiliated Xiangya Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Li Zhang
- Department of Engineering Physics, Tsinghua University Key Laboratory of Particle and Radiation Imaging, Ministry of Education, Haidian, Beijing 100084, P.R. China
| | - Yingrui Shi
- Department of Radiation Oncology, The Affiliated Hunan Provincial Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | | | - Guirong Wang
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Xiao Li
- Department of Radiation Oncology, The Affiliated Xiangya Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Weiwei Ouyang
- Department of Thoracic Oncology, Guizhou Cancer Hospital, Guiyang, Guizhou 550004, P.R. China
| | - Jumei Zhou
- Department of Radiation Oncology, The Affiliated Hunan Provincial Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Lingyun Zhao
- Department of Radiation Oncology, The Affiliated Xiangya Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Xiaowen Wang
- Department of Radiation Oncology, The Affiliated Xiangya Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Xiaodong Zhang
- Department of Radiation Oncology, The Affiliated Xiangya Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Fuping Gao
- Department of Radiation Oncology, The Affiliated Xiangya Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Jingshi Liu
- Department of Anesthesiology, Hunan Provincial Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, Hunan 410013, P.R. China
| | - Junming Luo
- Department of Radiation Oncology, The Affiliated Hunan Provincial Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China ; Department of Pathology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Jintian Tang
- Department of Radiation Oncology, The Affiliated Xiangya Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
30
|
Sharabi A, Herman J, Weiss V, Laheru D, Tuli R. Role of radiotherapy in combination with chemotherapy, targeted therapy, and immunotherapy in the management of pancreatic cancer. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s13566-013-0125-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
31
|
Kalbasi A, June CH, Haas N, Vapiwala N. Radiation and immunotherapy: a synergistic combination. J Clin Invest 2013; 123:2756-63. [PMID: 23863633 DOI: 10.1172/jci69219] [Citation(s) in RCA: 221] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy can be an effective treatment for metastatic cancer, but a significant subpopulation will not respond, likely due to the lack of antigenic mutations or the immune-evasive properties of cancer. Likewise, radiation therapy (RT) is an established cancer treatment, but local failures still occur. Clinical observations suggest that RT may expand the therapeutic reach of immunotherapy. We examine the immunobiologic and clinical rationale for combining RT and immunotherapy, two modalities yet to be used in combination in routine practice. Preclinical data indicate that RT can potentiate the systemic efficacy of immunotherapy, while activation of the innate and adaptive immune system can enhance the local efficacy of RT.
Collapse
Affiliation(s)
- Anusha Kalbasi
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | |
Collapse
|
32
|
Courtin A, Richards FM, Bapiro TE, Bramhall JL, Neesse A, Cook N, Krippendorff BF, Tuveson DA, Jodrell DI. Anti-tumour efficacy of capecitabine in a genetically engineered mouse model of pancreatic cancer. PLoS One 2013; 8:e67330. [PMID: 23840665 PMCID: PMC3696095 DOI: 10.1371/journal.pone.0067330] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/16/2013] [Indexed: 12/17/2022] Open
Abstract
Capecitabine (CAP) is a 5-FU pro-drug approved for the treatment of several cancers and it is used in combination with gemcitabine (GEM) in the treatment of patients with pancreatic adenocarcinoma (PDAC). However, limited pre-clinical data of the effects of CAP in PDAC are available to support the use of the GEMCAP combination in clinic. Therefore, we investigated the pharmacokinetics and the efficacy of CAP as a single agent first and then in combination with GEM to assess the utility of the GEMCAP therapy in clinic. Using a model of spontaneous PDAC occurring in Kras(G12D); p53(R172H); Pdx1-Cre (KPC) mice and subcutaneous allografts of a KPC PDAC-derived cell line (K8484), we showed that CAP achieved tumour concentrations (∼25 µM) of 5-FU in both models, as a single agent, and induced survival similar to GEM in KPC mice, suggesting similar efficacy. In vitro studies performed in K8484 cells as well as in human pancreatic cell lines showed an additive effect of the GEMCAP combination however, it increased toxicity in vivo and no benefit of a tolerable GEMCAP combination was identified in the allograft model when compared to GEM alone. Our work provides pre-clinical evidence of 5-FU delivery to tumours and anti-tumour efficacy following oral CAP administration that was similar to effects of GEM. Nevertheless, the GEMCAP combination does not improve the therapeutic index compared to GEM alone. These data suggest that CAP could be considered as an alternative to GEM in future, rationally designed, combination treatment strategies for advanced pancreatic cancer.
Collapse
Affiliation(s)
- Aurélie Courtin
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Frances M. Richards
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Tashinga E. Bapiro
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Jo L. Bramhall
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Albrecht Neesse
- Tumour Modelling and Experimental Medicine Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Natalie Cook
- Tumour Modelling and Experimental Medicine Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Ben-Fillippo Krippendorff
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - David A. Tuveson
- Tumour Modelling and Experimental Medicine Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Duncan I. Jodrell
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| |
Collapse
|
33
|
Kaplon R, Hadziahmetovic M, Sommerfeld J, Bondra K, Lu L, Leasure J, Nguyen P, McHugh K, Li N, Chronowski C, Sebastian N, Singh M, Kurmasheva R, Houghton P, Pelloski CE. The application of radiation therapy to the Pediatric Preclinical Testing Program (PPTP): results of a pilot study in rhabdomyosarcoma. Pediatr Blood Cancer 2013; 60:377-382. [PMID: 22692929 PMCID: PMC4733640 DOI: 10.1002/pbc.24210] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 05/07/2012] [Indexed: 11/08/2022]
Abstract
BACKGROUND The Pediatric Preclinical Testing Program (PPTP) has been successfully used to determine the efficacy of novel agents against solid tumors by testing them within a mouse-flank in vivo model. To date, radiation therapy has not been applied to this system. We report on the feasibility and biologic outcomes of a pilot study using alveolar and embryonal rhabdomyosarcoma xenograft lines. PROCEDURES We developed a high-throughput mouse-flank irradiation device that allows the safe delivery of radiotherapy in clinically relevant doses. For our pilot study, two rhabdomyosarcoma xenograft lines from the PPTP, Rh30 (alveolar) and Rh18 (embryonal) were selected. Using established methods, xenografts were implanted, grown to appropriate volumes, and were subjected to fractionated radiotherapy. Tumor response-rates, growth kinetics, and event-free survival time were measured. RESULTS Once optimized, the rate of acute toxicity requiring early removal from study in 93 mice was only 3%. During the optimization phase, it was observed that the alveolar Rh30 xenograft line demonstrated a significantly greater radiation resistance than embryonal Rh18 in vivo. This finding was validated within the standardized 30 Gy treatment phase, resulting in overall treatment failure rates of 10% versus 60% for the embryonal versus alveolar subtype, respectively. CONCLUSIONS Our pilot study demonstrated the feasibility of our device which enables safe, clinically relevant focal radiation delivery to immunocompromised mice. It further recapitulated the expected clinical radiobiology.
Collapse
Affiliation(s)
- Rita Kaplon
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | - Mersiha Hadziahmetovic
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | - Jim Sommerfeld
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | - Kathryn Bondra
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | - Lanchun Lu
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | - Justin Leasure
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | - Phuong Nguyen
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | - Kelsey McHugh
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | - Ning Li
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | - Christopher Chronowski
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | - Nikhil Sebastian
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | - Mamta Singh
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio
| | | | | | - Christopher E. Pelloski
- Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, Columbus, Ohio,Nationwide Children's Hospital, Columbus, Ohio,Correspondence to: Christopher E. Pelloski, MD, Wexner Medical Center at The Ohio State University, Arthur G. James Comprehensive Cancer Center and Richard L. Solove Research Institute, 300 West 10th Avenue, Suite 094A, Columbus, OH 43210-1280.
| |
Collapse
|
34
|
Kim ST, Lee J, Park SH, Lee JK, Lee KT, Lee KH, Heo JS, Choi SH, Choi DW, Park YS, Lim HY, Kang WK, Jang KT, Park HC, Lim DH, Park JO. Safety and efficacy of adjuvant chemoradiation therapy with capecitabine after resection of pancreatic ductal adenocarcinoma: a retrospective review. Am J Clin Oncol 2012; 35:432-438. [PMID: 21606820 DOI: 10.1097/coc.0b013e31821a83d7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To evaluate clinical outcomes and safety of adjuvant chemoradiation therapy (CRT) with capecitabine after resection of pancreatic adenocarcinoma at a single institution. PATIENTS AND METHODS A retrospective analysis of patients undergoing adjuvant CRT with capecitabine after resection of pancreatic ductal adenocarcinoma between 2004 and 2007 yielded a total of 55 patients. Capecitabine was administered at 850 mg/m(2) twice daily every day per week radiotherapy (45 Gy in 25 fractions) over the 5 weeks. Sixteen percent of patients (N=9) went on to receive gemcitabine. RESULTS Of 55 patients, 42 had curative (R0) resection and 13 had incomplete resection (R1). Median overall survival (OS) and progression free survival were 18.3 and 8.0 months for all patients, respectively. Patients receiving additional gemcitabine after adjuvant CRT with capecitabine showed better OS and progression free survival than those not receiving additional gemcitabine (P<0.05). In multivariate analysis, lymphovascular invasion (present vs. absent) and addition gemcitabine therapy (yes vs. no) were significant independent prognostic factors for OS (P<0.05). Local recurrence was observed in 10 patients, and distant recurrence in 26 patients, synchronously accounting for 6 recurrences. Ten patients (18.2%) had severe grade 3 toxicities. CONCLUSIONS Capecitabine-based CRT after resection of pancreatic adenocarcinoma showed favorable outcomes and tolerable toxicity profiles.
Collapse
Affiliation(s)
- Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Long J, Luo G, Liu C, Cui X, Satoh K, Xiao Z, Zhang B, Xu J, Ni Q, Li M, Yu X. Development of a unique mouse model for pancreatic cancer lymphatic metastasis. Int J Oncol 2012; 41:1662-8. [PMID: 22941445 DOI: 10.3892/ijo.2012.1613] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 07/20/2012] [Indexed: 11/06/2022] Open
Abstract
Lymphatic metastasis of pancreatic cancer is a predictor of poor prognosis. However, the molecular mechanisms are largely unknown, thus, making the development of appropriate cell lines and experimental models critically important for future investigations. The purpose of the present study was to establish a 'pancreatic cancer cell and mouse model with high lymphatic metastasis potential' for in-depth study of the underlying mechanisms. The BxPC-3-LN subline, derived from the BxPC-3 human pancreatic cancer cell line, was established through serial passages in nude mice via footpad injections. The subline was able to develop notable lymphatic metastases in 100% of the recipient mice 8 weeks after tumor cell implantation. Compared with the parental BxPC-3 cells, BxPC-3-LN cells were more aggressive, displaying invasive ultrastructure, increased migration and invasion ability, and chemoresistance. Metastasis-related gene alteration including upregulation of MMP14, MMP24, MIF and ADRM1, and downregulation of TGFB2 and ROBO1 were also observed in BxPC-3-LN cells by cDNA microarrays. Thus, the newly selected BxPC-3-LN subline can serve as a unique model for further study of lymphatic metastasis of pancreatic cancer.
Collapse
Affiliation(s)
- Jiang Long
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ma WW, Herman JM, Jimeno A, Laheru D, Messersmith WA, Wolfgang CL, Cameron JL, Pawlik TM, Donehower RC, Rudek MA, Hidalgo M. A tolerability and pharmacokinetic study of adjuvant erlotinib and capecitabine with concurrent radiation in resected pancreatic cancer. Transl Oncol 2010; 3:373-9. [PMID: 21151476 PMCID: PMC3000462 DOI: 10.1593/tlo.10196] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2010] [Revised: 08/25/2010] [Accepted: 08/26/2010] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Erlotinib is approved for the treatment of advanced pancreas cancer. We conducted a prospective trial to determine the safety profile and recommended phase 2 dose of erlotinib and capecitabine given concurrently with intensity-modulated radiation therapy (IMRT) in resected pancreatic cancer patients. The pharmacokinetic profile of this combination was also evaluated. METHODS Patients with resected pancreatic adenocarcinoma received erlotinib and capecitabine concurrently with IMRT delivered at 1.8 Gy daily in 28 fractions (total = 50.4 Gy). The starting dose level (DL 1) was erlotinib 150mgdaily and capecitabine 800 mg/m(2) twice daily without interruption. The next lower dose level (DL -1) was erlotinib 100 mg daily and capecitabine 800 mg/m(2) twice daily (Monday to Friday). Plasma samples were obtained for pharmacokinetic analysis. RESULTS Thirteen patients were enrolled in total. At DL 1, six of the seven treated patients were evaluable for toxicities. Four completed planned treatment, but all required treatment interruption or dose reduction. The dose-limiting toxicities were neutropenia, diarrhea, and rash. Six patients were subsequently enrolled to and completed planned treatment in DL-1. Themost common toxicities were fatigue, elevated liver enzymes, and anorexia. The pharmacokinetic parameters of erlotinib and OSI-420 were not significantly different in the presence or absence of capecitabine and were consistent with historical controls. CONCLUSIONS When administered concurrently with IMRT, erlotinib 100 mg daily and capecitabine 800 mg/m(2) twice daily (Monday to Friday) can be administered safely in resected pancreas cancer patients, and is the recommended regimen for efficacy studies using this regimen.
Collapse
Affiliation(s)
- Wen Wee Ma
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Strimpakos AS, Syrigos KN, Saif MW. The molecular targets for the diagnosis and treatment of pancreatic cancer. Gut Liver 2010; 4:433-49. [PMID: 21253292 PMCID: PMC3021599 DOI: 10.5009/gnl.2010.4.4.433] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2010] [Accepted: 10/18/2010] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer is considered an aggressive malignancy that responds poorly to current treatments and therefore has a dismal survival rate. This disease is usually not diagnosed until a late stage, at which point palliative chemotherapy with the purine analogue gemcitabine and/or a fluoropyrimidine or a platinum agent is the standard approach. There are some new data on the molecular and genetic changes that take place in pancreatic cancer, which may facilitate the accuracy of diagnosis and efficacy of treatments. However, translational efforts in clinical practice have increased clinicians' options with a targeted agent, erlotinib, in combination with the standard gemcitabine chemotherapy. Many other novel drugs currently being tested in the field of pharmaco-oncology target various altered biological pathways and molecules. Nevertheless, the lack of clinically significant improvements in treatments is rendering efforts to develop methods of early diagnosis both more urgent and promising. The aim of this review was to summarize the molecular basis of pancreatic carcinogenesis and the latest developments in diagnosis by molecular means, focusing on the results of clinical research into targeted and personalized treatments.
Collapse
Affiliation(s)
| | - Kostas N. Syrigos
- Oncology Unit, 3rd Department of Medicine, Sotiria General Hospital, Athens, Greece
| | - Muhammad Wasif Saif
- Division of Hematology/Oncology, Department of Medicine, Columbia University College of Physicians and Surgeons and Pancreas Center at the New York-Presbyterian Hospital, New York, NY, USA
| |
Collapse
|
38
|
Arjona-Sánchez A, Ruiz-Rabelo J, Perea MD, Vázquez R, Cruz A, Muñoz MDC, Túnez I, Muntané J, Padillo FJ. Effects of capecitabine and celecoxib in experimental pancreatic cancer. Pancreatology 2010; 10:641-7. [PMID: 21051919 DOI: 10.1159/000288708] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Accepted: 02/05/2010] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Pancreatic cancer is a major health problem because of its aggressiveness and the lack of effective systemic therapies. The aim of the study was the identification of beneficial properties of combined celecoxib and capecitabine treatment during an experimental pancreatic cancer model. METHODS N-nitrosobis (2-oxopropyl)amine (BOP) was used as a tumoral agent for 12 weeks. Celecoxib and capecitabine were administered either as monotherapy or combined 12 weeks after cancer induction for a period of 24 weeks. The presence of well-developed or moderate adenocarcinoma was evaluated in the pancreas. Several markers of stress, such as lipoperoxides, reduced glutathione (GSH), superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GHS-Px) were determined. RESULTS BOP induced the presence of pancreatic tumors associated with a rise in lipoperoxides and the reduction of the antioxidant status in the pancreas. The administration of celecoxib and capecitabine reduced the number of animals with tumors (33 and 66%, respectively). This antitumoral effect was associated with a recovery of GSH, SOD and CAT activity in the pancreas of BOP-treated animals. The combined treatment exerted a synergic antitumoral effect and reduced pancreatic oxidative stress. CONCLUSION The combined administration of celecoxib and capecitabine exerted a synergistic antitumoral effect and increased the antioxidant status restoration in pancreatic cancer.
Collapse
|
39
|
Di Gennaro E, Piro G, Chianese MI, Franco R, Di Cintio A, Moccia T, Luciano A, de Ruggiero I, Bruzzese F, Avallone A, Arra C, Budillon A. Vorinostat synergises with capecitabine through upregulation of thymidine phosphorylase. Br J Cancer 2010; 103:1680-91. [PMID: 21045833 PMCID: PMC2994231 DOI: 10.1038/sj.bjc.6605969] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background: Potentiation of anticancer activity of capecitabine is required to improve its therapeutic index. In colorectal cancer (CRC) cells, we evaluated whether the histone deacetylase-inhibitor vorinostat may induce synergistic antitumour effects in combination with capecitabine by modulating the expression of thymidine phosphorylase (TP), a key enzyme in the conversion of capecitabine to 5-florouracil (5-FU), and thymidylate synthase (TS), the target of 5-FU. Methods: Expression of TP and TS was measured by real-time PCR, western blotting and immunohistochemistry. Knockdown of TP was performed by specific small interfering RNA. Antitumour activity of vorinostat was assessed in vitro in combination with the capecitabine active metabolite deoxy-5-fluorouridine (5′-DFUR) according to the Chou and Talay method and by evaluating apoptosis as well as in xenografts-bearing nude mice in combination with capecitabine. Results: Vorinostat induced both in vitro and in vivo upregulation of TP as well as downregulation of TS in cancer cells, but not in ex vivo treated peripheral blood lymphocytes. Combined treatment with vorinostat and 5′-DFUR resulted in a synergistic antiproliferative effect and increased apoptotic cell death in vitro. This latter effect was impaired in cells where TP was knocked. In vivo, vorinostat plus capecitabine potently inhibited tumour growth, increased apoptosis and prolonged survival compared with control or single-agent treatments. Conclusions: Overall, this study suggests that the combination of vorinostat and capecitabine is an innovative antitumour strategy and warrants further clinical evaluation for the treatment of CRC.
Collapse
Affiliation(s)
- E Di Gennaro
- Experimental Pharmacology Unit, Department of Research, Istituto Nazionale Tumori, National Cancer Institute Fondazione G, Via M Semmola, Pascale, Napoli 80131, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bioevaluation of a novel [32P]-CP-PLLA microparticle for pancreatic cancer treatment. Drug Dev Res 2010. [DOI: 10.1002/ddr.20379] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
41
|
Unger KR, Romney DA, Koc M, Moskaluk CA, Friel CM, Foley EF, Rich TA. Preoperative chemoradiation for rectal cancer using capecitabine and celecoxib correlated with posttreatment assessment of thymidylate synthase and thymidine phosphorylase expression. Int J Radiat Oncol Biol Phys 2010; 80:1377-82. [PMID: 20656421 DOI: 10.1016/j.ijrobp.2010.04.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Revised: 04/19/2010] [Accepted: 04/19/2010] [Indexed: 11/27/2022]
Abstract
PURPOSE Thymidylate synthase (TS) and thymidine phosphorylase (TP) expression have been shown to be predictors of response to therapy. The toxicity, efficacy, surgical morbidity, and immunohistochemical TS and TP expression were assessed in surgical resection specimens after preoperative chemoradiation. METHODS AND MATERIALS Twenty patients with clinical stage I to III rectal adenocarcinoma received preoperative chemoradiation and underwent surgical resection 6 weeks later. RESULTS Posttreatment tumor stages were T1 to T2 and N0 in 30% of patients; T3 to T4 and N0 in 30% of patients; and T1 to T3 and N1 to N2 in 15% of patients. Pathologic complete response (pCR) was evident in 25% and tumor regression occurred in a total of 80% of patients. Anal sphincter-sparing surgery was performed in 80% of cases. Acute and perioperative complications were minimal, with no grade 3/4 toxicity or treatment breaks. Pelvic control was obtained in 90% of patients. With a median follow-up of 65.5 months (range, 8-80 months), the 6-year actuarial survival rate was 75%. Local failure was significantly associated with nonresponse to therapy and with high TS and low TP expression (p = 0.008 and p = 0.04, respectively). CONCLUSIONS The combination of capecitabine, celecoxib, and x-radiation therapy yields excellent response: a 25% pathologic pCR, no acute grade 3/4 toxicity, and minimal surgical morbidity. Nonresponders expressed significantly increased TS levels and decreased TP levels in posttreatment resection specimens compared to responders.
Collapse
Affiliation(s)
- Keith R Unger
- Department of Radiation Oncology, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Jackson ASN, Jain P, Watkins GR, Whitfield GA, Green MM, Valle J, Taylor MB, Dickinson C, Price PM, Saleem A. Efficacy and tolerability of limited field radiotherapy with concurrent capecitabine in locally advanced pancreatic cancer. Clin Oncol (R Coll Radiol) 2010; 22:570-7. [PMID: 20650619 DOI: 10.1016/j.clon.2010.06.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Revised: 03/22/2010] [Accepted: 05/11/2010] [Indexed: 11/19/2022]
Abstract
AIMS Patients with locally advanced pancreatic cancer (LAPC) are most commonly managed with chemotherapy or concurrent chemoradiotherapy (CRT), which may or may not include non-involved regional lymph nodes in the clinical target volume. We present our results of CRT for LAPC using capecitabine and delivering radiotherapy to a limited radiation field that excluded non-involved regional lymph nodes from the clinical target volume. MATERIALS AND METHODS Thirty patients were studied. Patients received 50.4 Gy external beam radiotherapy in 28 fractions, delivered to a planning target volume expanded from the primary tumour and involved nodes only. Capecitabine (500-600 mg/m2) was given twice daily continuously during radiotherapy. Toxicity and efficacy data were prospectively collected. RESULTS Nausea, vomiting and tumour pain were the most common grade 2 toxicities. One patient developed grade 3 nausea. The median time to progression was 8.8 months, with 20% remaining progression free at 1 year. The median overall survival was 9.7 months with a 1 year survival of 30%. Of 21 patients with imaged progression, 13 (62%) progressed systemically, three (14%) had local progression, two (10%) had locoregional progression and three (14%) progressed with both local/locoregional and systemic disease. CONCLUSION CRT using capecitabine and limited field radiotherapy is a well-tolerated, relatively efficacious treatment for LAPC. The low toxicity and low regional progression rates support the use of limited field radiotherapy, allowing evaluation of this regimen with other anti-cancer agents.
Collapse
Affiliation(s)
- A S N Jackson
- Academic Radiation Oncology, The University of Manchester, Department of Medical Oncology, The Christie Hospital NHS Foundation Trust, Manchester, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kim HS, Yi SY, Jun HJ, Lee J, Park SH, Lee JK, Lee KT, Lee KH, Choi DW, Choi SH, Heo JS, Park YS, Lim HY, Kang WK, Park HC, Lim DH, Park JO. Definitive chemoradiation therapy with capecitabine in locally advanced pancreatic cancer. Anticancer Drugs 2010; 21:107-112. [PMID: 19829097 DOI: 10.1097/cad.0b013e328332a7fc] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We evaluated safety and efficacy of concurrent chemoradiotherapy (CCRT) with capecitabine in patients with locally advanced pancreatic cancer (LAPC). Between January 2004 and January 2008, 39 patients with LAPC treated with capecitabine CCRT were reviewed. Capecitabine was administered at 850 mg/m twice daily every day with 5 days per week radiotherapy (1.8 Gy fractions) over the 5 weeks. Thirty-seven (94.8%) patients completed CCRT. Of the 36 evaluable patients, 15 (41.7%) and 13 (36.1%) patients achieved partial response and stable disease, and eight (28.6%) among them received gemcitabine-based post-CCRT chemotherapy without dose reduction or delay. The overall survival was 14.3 months [95% confidence interval (CI): 10.6-17.9 months]. Median progression-free survival was 11.1 months for all patients, and 7.9 months for those patients who had not received post-CCRT chemotherapy. Eight patients (21.6%) had severe grade 3 toxicities, seven (18.9%) with gastrointestinal toxicity, and one (2.7%) with hematologic toxicity. Prognostic factors for survival were serum albumin (P = 0.014; relative risk: 3.4; 95% CI: 1.4-9.7), and adjuvant gemcitabine treatment (P=0.005; relative risk: 3.5; 95% CI: 1.2-10.6). Combined therapy with capecitabine CCRT was well tolerated and seems to be a promising regimen, in terms of response, survival, and adverse effects.
Collapse
Affiliation(s)
- Hyo Song Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-dong, Kangnam-Gu, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Barrett-Lee P, Bidard FC, Pierga JY. Contemporary issues and the potential uses of capecitabine in metastatic breast cancer. Cancer Treat Rev 2009; 35:582-9. [PMID: 19632050 DOI: 10.1016/j.ctrv.2009.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 06/10/2009] [Accepted: 06/12/2009] [Indexed: 11/26/2022]
Abstract
Since its first regulatory approval more than 10 years ago, oncologists have gained wide experience in using the oral fluoropyrimidine, capecitabine, as monotherapy or in combination with other agents and the body of evidence supporting these approaches continues to grow. Alongside this increasing experience has been the appearance of new challenges in patient management. We now recognise several different biological subtypes of breast cancer, such as HER2-positive disease. The standard of care in these tumours comprises anti-HER2 therapy, and phase III data show that capecitabine can be effectively combined with such agents. Another increasingly prominent and currently unresolved issue resulting from more effective treatment of metastatic disease is the management of patients with brain metastases. The introduction of new, well-tolerated, oral chemotherapies also provides the opportunity for longer duration of therapy. These new clinical scenarios are discussed in the current review.
Collapse
|
45
|
Abstract
Radiotherapy is generally used to treat a localised target that includes cancer. Increasingly, evidence indicates that radiotherapy recruits biological effectors outside the treatment field and has systemic effects. We discuss the implications of such effects and the role of the immune system in standard cytotoxic treatments. Because the effects of chemotherapy and radiotherapy are sensed by the immune system, their combination with immunotherapy presents a new therapeutic opportunity. Radiotherapy directly interferes with the primary tumour and possibly reverses some immunosuppressive barriers within the tumour microenvironment-ideally, recovering the role of the primary tumour as an immunogenic hub. Local radiation also triggers systemic effects that can be used in combination with immunotherapy to induce responses outside the radiation field.
Collapse
Affiliation(s)
- Silvia C Formenti
- Department of Radiation Oncology, NYU Langone Medical Center and NYU Cancer Institute, New York University School of Medicine, New York, NY 10016, USA.
| | | |
Collapse
|
46
|
Johansson AS, Pawelzik SC, Larefalk A, Jakobsson PJ, Holmberg D, Lindskog M. Lymphoblastic T-cell lymphoma in mice is unaffected by Celecoxib as single agent or in combination with cyclophosphamide. Leuk Lymphoma 2009; 50:1198-203. [PMID: 19557641 DOI: 10.1080/10428190902946930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Celecoxib, an inhibitor of cyclooxygenase-2, is a promising novel antitumor agent with pleitropic mechanisms of action. Whereas this drug induces growth arrest and apoptosis of B-lymphoma cells, its effect against aggressive T-cell neoplasms remains to be studied. We therefore evaluated Celecoxib therapy of immunocompetent mice transplanted with lymphoblastic T-cell lymphomas. Oral Celecoxib in clinically relevant and non-toxic doses did not affect the degree of hypersplenism or the number of viable lymphoma cells. The clinical deterioration of Celecoxib-treated mice was not different from untreated controls. The impact of adding Celecoxib (60 mg/kg) to cyclophosphamide (200 mg/kg x 1, i.p.) was assessed but showed no benefit compared to cyclophosphamide alone. Thus, Celecoxib lacks effect against lymphoblastic T-cell lymphoma in mice.
Collapse
|
47
|
Strimpakos A, Saif MW, Syrigos KN. Pancreatic cancer: from molecular pathogenesis to targeted therapy. Cancer Metastasis Rev 2008; 27:495-522. [PMID: 18427734 DOI: 10.1007/s10555-008-9134-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pancreatic cancer is a deadly malignancy with still high mortality and poor survival despite the significant advances in understanding, diagnosis, and access to conventional and novel treatments. Though cytotoxic chemotherapy based on the purine analogue gemcitabine remains the standard approach in adjuvant and palliative setting the need for novel agents aiming at the main pathophysiological abnormalities and molecular pathways involved remains soaring. So far, evidence of clinical benefit, though small, exists only from the addition of the targeted agent erlotinib on the standard gemcitabine chemotherapy. Apart from the popular monoclonal antibodies and small molecules tyrosine kinase inhibitors, other novel compounds being tested in preclinical and clinical studies target mTOR, NF-kappaB, proteasome and histone deacetylase. These new drugs along with gene therapy and immunotherapy, which are also under clinical evaluation, may alter the unfavorable natural course of this disease. In this review we present the main pathophysiological alterations met in pancreatic cancer and the results of the florid preclinical and clinical research with regards to the targeted therapy associated to these abnormalities.
Collapse
|
48
|
Riess H. Antiangiogenic strategies in pancreatic cancer. RECENT RESULTS IN CANCER RESEARCH. FORTSCHRITTE DER KREBSFORSCHUNG. PROGRES DANS LES RECHERCHES SUR LE CANCER 2007; 177:123-9. [PMID: 18084954 DOI: 10.1007/978-3-540-71279-4_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Since the introduction of gemcitabine in the treatment of pancreatic cancer, progress in the use of combination chemotherapies has been very limited. Of the different novel options, antiangiogenic treatment strategies are among those being intensively studied in preclinical and clinical settings of adenocarcinoma of the pancreas. Phase I and limited-size phase II studies using drugs with antiangiogenic properties have reported encouraging results. Overall, the results of phase III studies with some metalloprotease inhibitors and bevacizumab have so far failed to demonstrate a survival benefit for these drugs. Further investigations that will take into account the heterogeneity of pancreatic cancer are warranted using these or other antiangiogenic active substances.
Collapse
Affiliation(s)
- H Riess
- Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Medizinische Klinik mit Schwerpunkt Hämatologie und Onkologie, Germany
| |
Collapse
|
49
|
Saif MW, Black G, Roy S, Bell D, Russo S, Eloubeidi MA, Steg A, Johnson MR, Zelterman D, Diasio RB. Phase II study of capecitabine with concomitant radiotherapy for patients with locally advanced pancreatic cancer: up-regulation of thymidine phosphorylase. Cancer J 2007; 13:247-56. [PMID: 17762760 DOI: 10.1097/ppo.0b013e31813c12b8] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE The objectives of this phase II study were to evaluate the effect of radiation (XRT) on thymidine phosphorylase (TP), dihydropyrimidine dehydrogenase (DPD), and tumor necrosis factor-alpha (TNF-alpha) and the efficacy of capecitabine-XRT in patients with locally advanced pancreatic cancer. PATIENTS AND METHODS Twenty patients received 50.4 Gy XRT with capecitabine 1,600 mg/m(2) on Monday through Friday for 6 weeks determined from our phase I study (Saif MW, Eloubeidi MA, Russo S, et al. J Clin Oncol. 2005;23:8679-8687). After capecitabine-XRT, stable and responding patients received capecitabine 2,000 mg/m(2) for 14 days every 3 weeks till progression. Restaging was performed every 9 weeks. Tumor specimens were procured with endoscopic ultrasound-fine needle aspiration before and at week 2 after capecitabine-XRT was started to evaluate the effect of XRT on TP, DPD, and TNF-alpha mRNA levels, determined by reverse transcriptase-polymerase chain reaction. RESULTS Among 20 patients, 4 (20%) had a partial response and 13 (65%) had stable disease. Two patients underwent surgical resection (10%). The 6-month survival rate was 84%, and the 1-year survival was 58%. Grade > or =3 toxicities included nausea/vomiting (5%), thrombosis (5%), hyperbilirubinemia (5%), and grade 3 gastrointestinal bleeding (5%). TP was elevated during week 2 when compared with the pre-XRT TP (P = 0.01). However, no such effect of XRT was found either on DPD (P = 0.22) or on TNF-alpha (P = 0.6). No correlation between TP and TNF-alpha was noticed. Also, no association between TP/DPD ratio and efficacy of capecitabine was identified. CONCLUSIONS This phase II study further confirms our phase I results and suggests that capecitabine-XRT is an effective, tolerable, and convenient alternative to an infusional 5-fluorouracil regimen for patients with pancreatic cancer. Although results support the use of capecitabine-XRT and TP was up-regulated, there appears to be additional genes associated with the response to capecitabine.
Collapse
Affiliation(s)
- Muhammad Wasif Saif
- Departments of Medical Oncology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fanciullino R, Giacometti S, Mercier C, Aubert C, Blanquicett C, Piccerelle P, Ciccolini J. In vitro and in vivo reversal of resistance to 5-fluorouracil in colorectal cancer cells with a novel stealth double-liposomal formulation. Br J Cancer 2007; 97:919-26. [PMID: 17848948 PMCID: PMC2360412 DOI: 10.1038/sj.bjc.6603970] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Drug resistance is a major cause of treatment failure in cancer chemotherapy, including that with the extensively prescribed antimetabolite, 5-fluorouracil (5-FU). In this study, we tried to reverse 5-FU resistance by using a double-punch strategy: combining 5-FU with a biochemical modulator to improve its tumoural activation and encapsulating both these agents in one same stealth liposome. Experiments carried out in the highly resistant, canonical SW620 human colorectal model showed a up to 80% sensitisation to 5-FU when these cells were treated with our liposomal formulation. Results with this formulation demonstrated 30% higher tumoural drug uptake, better activation with increased active metabolites including critical-5-fluoro-2-deoxyuridine-5-monophosphate, superior inhibition (98%) of tumour thymidylate synthase, and subsequently, higher induction of both early and late apoptosis. Drug monitoring showed that higher and sustained exposure was achieved in rats treated with liposomal formulation. When examined in a xenograft animal model, our dual-agent liposomal formulation caused a 74% reduction in tumour size with a mean doubling in survival time, whereas standard 5-FU failed to exhibit significant antiproliferative activity as well as to increase the lifespan of tumour-bearing mice. Taken collectively, our data suggest that resistance to 5-FU can be overcome through a better control of its intratumoural activation and the use of an encapsulated formulation.
Collapse
Affiliation(s)
- R Fanciullino
- EA3286-Laboratoire de Pharmacocinétique, Université de la Méditerranée, Marseille, France
| | - S Giacometti
- EA3286-Laboratoire de Pharmacocinétique, Université de la Méditerranée, Marseille, France
| | - C Mercier
- EA3286-Laboratoire de Pharmacocinétique, Université de la Méditerranée, Marseille, France
| | - C Aubert
- EA3286-Laboratoire de Pharmacocinétique, Université de la Méditerranée, Marseille, France
| | - C Blanquicett
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - P Piccerelle
- Laboratoire de Pharmacie Galénique, Faculté de Pharmacie, 27 Bd Jean Moulin, Marseille 05 13385, France
| | - J Ciccolini
- EA3286-Laboratoire de Pharmacocinétique, Université de la Méditerranée, Marseille, France
- EA3286, Laboratoire de Pharmacocinétique, Faculté de Pharmacie, 27 Bd Jean Moulin 13385, Marseille 05, France. E-mail:
| |
Collapse
|