1
|
Zhang Z, Lu T, Zhang Z, Liu Z, Qian R, Qi R, Zhou F, Li M. Unraveling the immune landscape and therapeutic biomarker PMEPA1 for oxaliplatin resistance in colorectal cancer: A comprehensive approach. Biochem Pharmacol 2024; 222:116117. [PMID: 38461903 DOI: 10.1016/j.bcp.2024.116117] [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: 10/24/2023] [Revised: 02/20/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
Oxaliplatin (OXA) is a platinum-based chemotherapeutic agent with promising applications in the treatment of various malignancies, particularly colorectal cancer (CRC). However, the management of OXA resistance remains an ongoing obstacle in CRC therapy. This study aims to comprehensively investigate the immune landscape, targeted therapeutic biomarkers, and mechanisms that influence OXA resistance in CRC. Our results demonstrated that our OXA- resistant CRC prognostic model not only provides risk assessment for patients but also reflects the immune landscape of patients. Additionally, we identified prostate transmembrane protein, androgen-induced1 (PMEPA1) as a promising molecular targeted therapeutic biomarker for patients with OXA-resistant CRC. The mechanism of PMEPA1 may involve cell adhesion, pathways in cancer, and the TGF-β signaling pathway. Furthermore, analysis of CRC clinical samples indicated that patients resistant to OXA exhibited elevated serum levels of TGF-β1, increased expression of PMEPA1 in tumors, a lower proportion of CD8+ T cell positivity, and a higher proportion of M0 macrophage positivity, in comparison to OXA-sensitive individuals. Cellular experiments indicated that selective silencing of PMEPA1, alone or in combination with OXA, inhibited proliferation and metastasis in OXA-resistant CRC cells, HCT116R. Animal experiments further confirmed that PMEPA1 silencing suppressed subcutaneous graft tumor growth and liver metastasis in mice bearing HCT116R and synergistically enhanced the efficacy of OXA. These data highlight the potential of leveraging the therapeutic biomarker PMEPA1, CD8+ T cells, and M0 macrophages as innovative targets for effectively addressing the challenges associated with OXA resistance. Our findings hold promising implications for further clinical advancements in this field.
Collapse
Affiliation(s)
- Zhengguang Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| | - Tianming Lu
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Zhe Zhang
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Zixian Liu
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Ruoning Qian
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Ruogu Qi
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| | - Fuqiong Zhou
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| | - Min Li
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| |
Collapse
|
2
|
Lenz HJ, Parikh A, Spigel DR, Cohn AL, Yoshino T, Kochenderfer M, Elez E, Shao SH, Deming D, Holdridge R, Larson T, Chen E, Mahipal A, Ucar A, Cullen D, Baskin-Bey E, Kang T, Hammell AB, Yao J, Tabernero J. Modified FOLFOX6 plus bevacizumab with and without nivolumab for first-line treatment of metastatic colorectal cancer: phase 2 results from the CheckMate 9X8 randomized clinical trial. J Immunother Cancer 2024; 12:e008409. [PMID: 38485190 PMCID: PMC10941175 DOI: 10.1136/jitc-2023-008409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Standard first-line therapies for metastatic colorectal cancer (mCRC) include fluoropyrimidine-containing regimens with oxaliplatin and/or irinotecan and a biologic agent. Immunotherapy may enhance antitumor activity in combination with standard therapies in patients with mCRC. Here, we present phase 2 results of nivolumab plus standard-of-care therapy (SOC; 5-fluorouracil/leucovorin/oxaliplatin/bevacizumab) versus SOC in the first-line treatment of patients with mCRC (CheckMate 9X8). METHODS CheckMate 9X8 was a multicenter, open-label, randomized, phase 2/3 trial. Eligible patients were at least 18 years of age with unresectable mCRC and no prior chemotherapy for metastatic disease. Patients were randomized 2:1 to receive nivolumab 240 mg plus SOC or SOC alone every 2 weeks. The primary endpoint was progression-free survival (PFS) by blinded independent central review (BICR) per Response Evaluation Criteria in Solid Tumors V.1.1. Secondary endpoints included PFS by investigator assessment; objective response rate (ORR), disease control rate, duration of response, and time to response, all by BICR and investigator assessments; overall survival; and safety. Preplanned exploratory biomarker analyses were also performed. RESULTS From February 2018 through April 2019, 310 patients were enrolled, of which 195 patients were randomized to nivolumab plus SOC (n=127) or SOC (n=68). At 21.5-month minimum follow-up, PFS with nivolumab plus SOC versus SOC did not meet the prespecified threshold for statistical significance; median PFS by BICR was 11.9 months in both arms (HR, 0.81 (95% CI, 0.53 to 1.23); p=0.30). Higher PFS rates after 12 months (18 months: 28% vs 9%), higher ORR (60% vs 46%), and durable responses (median 12.9 vs 9.3 months) were observed with nivolumab plus SOC versus SOC. Grade 3-4 treatment-related adverse events were reported in 75% versus 48% of patients; no new safety signals were identified. CONCLUSIONS The CheckMate 9X8 trial investigating first-line nivolumab plus SOC versus SOC in patients with mCRC did not meet its primary endpoint of PFS by BICR. Nivolumab plus SOC showed numerically higher PFS rates after 12 months, a higher response rate, and more durable responses compared with SOC alone, with acceptable safety. Further investigation to identify subgroups of patients with mCRC that may benefit from nivolumab plus SOC versus SOC in the first-line setting is warranted. TRIAL REGISTRATION NUMBER NCT03414983.
Collapse
Affiliation(s)
- Heinz-Josef Lenz
- Department of Medical Oncology, USC Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Aparna Parikh
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David R Spigel
- Department of Oncology, Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | - Allen L Cohn
- Department of Medical Oncology, US Oncology Research, Rocky Mountain Cancer Centers, Denver, Colorado, USA
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center-Hospital East, Kashiwa, Chiba, Japan
| | | | - Elena Elez
- Department of Medical Oncology, Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Dustin Deming
- Departments of Medicine and Oncology, University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Regan Holdridge
- Comprehensive Cancer Centers of Nevada, Henderson, Nevada, USA
| | - Timothy Larson
- Department of Medical Oncology, Minnesota Oncology Hematology, Minneapolis, Minnesota, USA
| | - Eric Chen
- Department of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Amit Mahipal
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Antonio Ucar
- Miami Cancer Institute (part of Baptist Health South Florida), Miami, Florida, USA
| | - Dana Cullen
- Oncology Clinical Science, Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Tong Kang
- Biostatistics, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Amy B Hammell
- Precision Medicine, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Jin Yao
- Translational Bioinformatics, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Josep Tabernero
- Department of Medical Oncology, Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| |
Collapse
|
3
|
Silva TH, Sillos André JC, Orlando Correa Schilithz A, Borges Murad L, Arantes Ferreira Peres W. Prediction of survival of preoperative colorectal patients: A new tool to assess the interaction of nutritional status and inflammation. Clin Nutr ESPEN 2023; 56:230-236. [PMID: 37344078 DOI: 10.1016/j.clnesp.2023.05.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is responsible for the second leading cause of cancer death worldwide. Thus, the aim of this study was to investigate the application of a new nutritional status and inflammatory tool to predict overall survival (OS) in patients with CRC in the preoperative period. METHODS Retrospective cohort study with CRC patients of both sexes treated at a hospital unit, aged ≥20 years. Data were extracted between 2007 and 2015. Multivariate Cox's models were employed to predict OS utilizing a new grade classification system (body mass index vs. percentage weight loss - BMI/%WL), inflammatory markers and clinical data. RESULTS We evaluated 361 patients. BMI/%WL grade 3-4 (HR: 2.01; p = 0.001; 95% CI: 1.34-3.02) were independent predictors of poor OS. Moreover, BMI/%WL grade 3-4 + neutrophil-to-lymphocyte ratio (NLR) ≥2.4 (HR: 2.79; p = 0.001; 95% CI: 1.54-5.03) increased the death risk in 5-years. However, low NLR (<2.4) altered the OS prognostic ability of the BMI/%WL (HR: 1.72; p = 0.099; 95% CI: 0.90-3.28). CONCLUSION BMI/%WL was independent predictors of poor OS and the interaction with NLR produced an adjustment effect. These associated tools may be useful in the clinical management of preoperative patients with CRC.
Collapse
Affiliation(s)
| | - Julio Cezar Sillos André
- Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil; National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | | | | | | |
Collapse
|
4
|
Joshi D, Khursheed R, Gupta S, Wadhwa D, Singh TG, Sharma S, Porwal S, Gauniyal S, Vishwas S, Goyal S, Gupta G, Eri RD, Williams KA, Dua K, Singh SK. Biosimilars in Oncology: Latest Trends and Regulatory Status. Pharmaceutics 2022; 14:pharmaceutics14122721. [PMID: 36559215 PMCID: PMC9784530 DOI: 10.3390/pharmaceutics14122721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/07/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Biologic-based medicines are used to treat a variety of diseases and account for around one-quarter of the worldwide pharmaceutical market. The use of biologic medications among cancer patients has resulted in substantial advancements in cancer treatment and supportive care. Biosimilar medications (or biosimilars) are very similar to the reference biologic drugs, although they are not identical. As patent protection for some of the most extensively used biologics begins to expire, biosimilars have the potential to enhance access and provide lower-cost options for cancer treatment. Initially, regulatory guidelines were set up in Europe in 2003, and the first biosimilar was approved in 2006 in Europe. Many countries, including the United States of America (USA), Canada, and Japan, have adopted Europe's worldwide regulatory framework. The use of numerous biosimilars in the treatment and supportive care of cancer has been approved and, indeed, the count is set to climb in the future around the world. However, there are many challenges associated with biosimilars, such as cost, immunogenicity, lack of awareness, extrapolation of indications, and interchangeability. The purpose of this review is to provide an insight into biosimilars, which include various options available for oncology, and the associated adverse events. We compare the regulatory guidelines for biosimilars across the world, and also present the latest trends and challenges in medical oncology both now and in the future, which will assist healthcare professionals, payers, and patients in making informed decisions, increasing the acceptance of biosimilars in clinical practice, increasing accessibility, and speeding up the health and economic benefits associated with biosimilars.
Collapse
Affiliation(s)
- Deeksha Joshi
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India
| | - Rubiya Khursheed
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Saurabh Gupta
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India
| | - Diksha Wadhwa
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India
| | | | - Sumit Sharma
- Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Sejal Porwal
- Department of Pharmaceutical Sciences, Amity University Lucknow, Lucknow 226028, India
| | - Swati Gauniyal
- Department of Pharmacology, KLE College of Pharmacy, Hubballi 580031, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Sanjay Goyal
- Department of Internal Medicine, Government Medical College, Patiala 147001, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura 333031, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602117, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun 248007, India
| | - Rajaraman D. Eri
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
- Correspondence: (R.D.E.); (S.K.S.); Tel.: +61-3-6324-5467 (R.D.E.); +91-9888720835 (S.K.S.)
| | - Kylie A. Williams
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Correspondence: (R.D.E.); (S.K.S.); Tel.: +61-3-6324-5467 (R.D.E.); +91-9888720835 (S.K.S.)
| |
Collapse
|
5
|
Wang Y, Ma J, Qiu T, Tang M, Zhang X, Dong W. In vitro and in vivo combinatorial anticancer effects of oxaliplatin- and resveratrol-loaded N,O-carboxymethyl chitosan nanoparticles against colorectal cancer. Eur J Pharm Sci 2021; 163:105864. [PMID: 33965502 DOI: 10.1016/j.ejps.2021.105864] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 04/11/2021] [Accepted: 04/25/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Oxaliplatin (OXE) combined with other chemotherapy drugs against colorectal cancer had been reported in the literature before, however, the efficacy of oxaliplatin combined with natural compounds was elusive. In addition, the clinical bioactivity and therapeutic dose of antitumor drugs are severely limited due to poor targeting and side effects. NDDSs offers an excellent strategy to overcome the disadvantages of small molecule anticancer drugs. METHODS Here, we have prepared N,O-carboxymethyl chitosan Oxaliplatin nanoparticles (CMCS-OXE NPs) and N,O-carboxymethyl chitosan Resveratrol nanoparticles (CMCS-Res NPs) were prepared by ion crosslinking and emulsification crosslinking, respectively. RESULTS The results revealed that the CMCS-OXE NPs exhibited a high encapsulation efficiency (60%) with a size of approximately 190.0 nm, and the CMCS-Res NPs exhibited a high encapsulation efficiency (65%) with a size of approximately 164.2 nm. The treatment with both types of nanoparticles combined exhibited more significant anti-colon cancer activity than the free drugs or either type of nanoparticle alone. In the in vivo experiments, the inhibition efficiency of the combined nanoparticle treatment was much stronger than the free drugs or either type of nanoparticle alone. CONCLUSIONS Overall, combination of oxaliplatin and resveratrol into a nanoparticle-drug delivery systems (NDDSs) appears to be a promising strategy for colorectal cancer (CRC) therapy.
Collapse
Affiliation(s)
- Yaowen Wang
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China.
| | - Jingjing Ma
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430070, China; Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, Wuhan 430060, China.
| | - Tong Qiu
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, China; State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China.
| | - Mingxiu Tang
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China.
| | - Xueqiong Zhang
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China.
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430070, China.
| |
Collapse
|
6
|
李 萍, 袁 平, 阙 月, 刘 筱, 王 国. [Synergistic effect of polysaccharide from Trichoderma pseudokoningii and oxaliplatin on colorectal cancer cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:504-513. [PMID: 33963708 PMCID: PMC8110445 DOI: 10.12122/j.issn.1673-4254.2021.04.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To explore the synergistic inhibitory effect of polysaccharide from Trichoderma pseudokoningii (EPS) and oxaliplatin (Oxa) on colorectal cancer (CRC) HCT116 cells. OBJECTIVE HCT116 cells were treated with 8 μg/mL Oxa and 100 μg/mL EPS alone or in combination, and the changes in cell viability was assessed with CCK-8 assay. CompuSyn software was used for fitting the Fa-CI curve to evaluate the combined effect of the two agents. Flow cytometry was performed to analyze cell apoptosis and cell cycle changes, and wound healing assay and Transwell assay were used to examine the migration ability of the treated cells. Oxa- and EPS-related genes and CRC-related genes were intersected for protein-protein interaction (PPI) analysis and GO and KEGG enrichment analyses. OBJECTIVE Treatment with Oxa alone or in combination with EPS significantly inhibited the viability of HCT116 cells in a dose- and time-dependent manner, and the two agents exhibited a significant synergistic effect (CI < 1). The combined treatment with Oxa and EPS resulted in a significantly higher total cell apoptosis rate and a higher percentage of cells in S phase than Oxa alone and the control treatment (P < 0.05). EPS and Oxa alone both inhibited the migration of HCT116 cells, and their combination produced a stronger inhibitory effect. GO enrichment analysis of the key genes related with Oxa, EPS and CRC suggested that these genes were involved mainly in such biological processes as exogenous apoptosis signaling, cell response to chemical stress, and reactive oxygen metabolism; KEGG analysis showed that these genes were involved in the pathways of drug resistance, apoptosis and angiogenesis. OBJECTIVE EPS and Oxa can synergistically inhibit the proliferation of HCT116 cells possibly through the PI3K-Akt, MAPK, VEGF, and p53 signaling pathways.
Collapse
Affiliation(s)
- 萍 李
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002School of Pharmacy, Wannan Medical College, Wuhu 241002, China
- 安徽省多糖药物工程技术研究中心//活性生物大分子研究安徽省重点实验室,安徽 芜湖 241002Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wuhu 241002, China
| | - 平川 袁
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002School of Pharmacy, Wannan Medical College, Wuhu 241002, China
- 安徽省多糖药物工程技术研究中心//活性生物大分子研究安徽省重点实验室,安徽 芜湖 241002Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wuhu 241002, China
| | - 月月 阙
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - 筱琴 刘
- 重庆化工职业学院//制药领域关键共性工艺重庆市高等职业技术院校应用技术推广中心,重庆 401220Chongqing Chemical Industry Vocational College, Chongqing Municipal Vocational and Technical College Application Technology Promotion Center for Key Common Technology in Pharmaceuticals, Chongqing 401220, China
| | - 国栋 王
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002School of Pharmacy, Wannan Medical College, Wuhu 241002, China
- 安徽省多糖药物工程技术研究中心//活性生物大分子研究安徽省重点实验室,安徽 芜湖 241002Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wuhu 241002, China
| |
Collapse
|
7
|
Nilsson S, Stein A, Rolfo C, Kranich AL, Mann J, Papadimitriou K, Theile S, Amberg S, Bokemeyer C. Selinexor (KPT-330), an Oral Selective Inhibitor of Nuclear Export (SINE) Compound, in Combination with FOLFOX in Patients with Metastatic Colorectal Cancer (mCRC) - Final Results of the Phase I Trial SENTINEL. Curr Cancer Drug Targets 2020; 20:811-817. [PMID: 32598257 DOI: 10.2174/1568009620666200628105727] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Selinexor is an oral Selective Inhibitor of Nuclear Export compound that specifically blocks Chromosomal Region Maintenance protein 1. OBJECTIVE To evaluate the safety and tolerability of escalating doses of selinexor plus 5-fluorouracil, leucovorin and oxaliplatin (mFOLFOX6) in metastatic colorectal cancer (mCRC) patients. METHODS In this multicenter phase I trial, mCRC patients, eligible for oxaliplatin-based treatment, were enrolled to receive oral selinexor on days 1, 3, and 8 plus mFOLFOX6 every two weeks. Primary endpoint was the maximum tolerated dose. Secondary endpoints were toxicity, overall response rate, progression free survival, and overall survival. RESULTS Overall, 10 patients were enrolled, who had prior treatment with oxaliplatin (6/10), irinotecan (8/10), bevacizumab (6/10) or anti-EGFR therapy (5/10). Four consecutive patients received 40 mg selinexor plus mFOLFOX6. All four experienced dose-limiting toxicities and withdrew from the study after a median of two cycles. Thus, this dose level was regarded as toxic and no further patients were evaluated at this dose. Six patients were enrolled with 20 mg selinexor plus mFOLFOX6. Despite better tolerability, four patients withdrew (patient wish) after the first cycle and only two patients continued until disease progression. Most commonly reported treatment emergent adverse events were nausea (80%), diarrhea (70%), vomiting (60%), fatigue (60%), anorexia (40%), and impaired vision (40%). Due to the short treatment exposure, no relevant clinical activity was observed. CONCLUSION In patients with metastatic colorectal cancer, selinexor on this dose schedule plus mFOLFOX6 was not tolerable. Other dosing schedules or combinations may be evaluated. Clinical trial identifier NCT02384850.
Collapse
Affiliation(s)
- Sven Nilsson
- II. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander Stein
- II. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Hematology-Oncology Practice Hamburg-Eppendorf (HOPE), Hamburg, Germany
| | - Christian Rolfo
- Phase I- Early Clinical Trials Unit, Antwerp University Hospital, Edegem, Belgium,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, United States of America
| | - Anne L Kranich
- GSO Global Clinical Research B.V., Amsterdam, the Netherlands
| | - Julia Mann
- II. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Susann Theile
- GSO Gesellschaft für Studienmanagement und Onkologie mbH, Hamburg, Germany
| | - Stefanie Amberg
- GSO Gesellschaft für Studienmanagement und Onkologie mbH, Hamburg, Germany
| | - Carsten Bokemeyer
- II. Medical Clinic and Polyclinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
8
|
Konstantinidou S, Papaspiliou A, Kokkotou E. Current and future roles of biosimilars in oncology practice. Oncol Lett 2019; 19:45-51. [PMID: 31897113 PMCID: PMC6923870 DOI: 10.3892/ol.2019.11105] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022] Open
Abstract
Biologics have been used increasingly in the treatment and supportive care of cancer; however, their high cost places a significant burden on healthcare systems. The expiration of patents for biologics has led to the development of biosimilars, with the aim of reducing cost and increasing accessibility to novel treatments, which are affordable for a greater number of patients. Biosimilars are highly similar but not identical to the reference products; therefore, strict regulatory requirements have been formed for their approval. This ensures that there are no clinically meaningful differences compared with respective biologics, with regard to purity, safety and efficacy. In 2003, a regulatory framework for the approval of biosimilars was established in Europe, whereas the USA did not implement a framework until 2009, when the Biologics Price Competition and Innovation Act was formed. A number of biosimilars have currently been approved in oncology and the number is expected to rise in the near future. More than 10 years of evidence has revealed that biosimilars are safe and effective; however healthcare professionals need to be further educated to eliminate potential misconceptions and integrate biosimilars into routine clinical practice. The present review aims to provide an overview of the biosimilars used in Europe and the USA, present their main benefits and challenges, and discuss their current and future roles in medical oncology.
Collapse
Affiliation(s)
- Sofia Konstantinidou
- Oncology Unit, The Third Department of Medicine, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Angeliki Papaspiliou
- Oncology Unit, The Third Department of Medicine, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Eleni Kokkotou
- Oncology Unit, The Third Department of Medicine, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| |
Collapse
|
9
|
Lu Y, Xin D, Wang F. Predictive Significance Of Preoperative Systemic Immune-Inflammation Index Determination In Postoperative Liver Metastasis Of Colorectal Cancer. Onco Targets Ther 2019; 12:7791-7799. [PMID: 31571929 PMCID: PMC6759789 DOI: 10.2147/ott.s223419] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose Systemic inflammation and immune dysfunction have been proved to be significantly associated with cancer progression and metastasis in colorectal cancer (CRC). The aim of this retrospective study was to investigate the association between preoperative systemic immune-inflammation index (SII) and postoperative liver metastasis in CRC. Patients and methods This retrospective study evaluated 182 patients with CRC who underwent surgical resection. The inflammation-based prognostic factors, including SII, neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), lymphocyte to monocyte ratio (LMR) and prognostic nutritional index (PNI), were calculated based on preoperative laboratory data. The univariate and multivariate logistic regression analysis was performed to identify the risk factors correlated with postoperative liver metastasis in CRC. Receiver operating characteristic (ROC) curves and decision curve analysis (DCA) were respectively used to assess the predictive ability and clinical usefulness of SII for postoperative liver metastasis in CRC. Results The univariate and multivariable analysis confirmed SII was independently correlated with postoperative liver metastasis in CRC (p<0.001), and the ROC and DCA analysis demonstrated SII was superior to other inflammation-based factors in terms of predictive ability. Conclusion SII is an independent predictive indicator of postoperative liver metastasis for patients with colorectal cancer.
Collapse
Affiliation(s)
- Yao Lu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Dao Xin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| |
Collapse
|
10
|
Deng XQ, Zhang HB, Wang GF, Xu D, Zhang WY, Wang QS, Cui YL. Colon-specific microspheres loaded with puerarin reduce tumorigenesis and metastasis in colitis-associated colorectal cancer. Int J Pharm 2019; 570:118644. [PMID: 31465837 DOI: 10.1016/j.ijpharm.2019.118644] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/06/2019] [Accepted: 08/24/2019] [Indexed: 02/07/2023]
Abstract
Colitis-associated colorectal cancer (CAC) is a common malignancy that develops in chronically inflamed mucosa and is usually accompanied by metastases at other sites. Puerarin, a natural isoflavone isolated from the root of the Pueraria lobata (Willd.) Ohwi, has potential anti-colon cancer activity. However, the poor solubility and low bioavailability of puerarin has restricted its application in the pharmaceutical industry. In the present study, pH-responsive alginate microspheres loaded with puerarin were prepared by emulsification/internal gelation for targeted treatment of colitis-associated colorectal cancer. Herein, puerarin, as an active drug, could participate in the construction of alginate microspheres with hydrogen bonding. The microspheres exhibited pH-responsive release behavior with little release of puerarin in simulated gastric fluid and high amounts (approximately 55%) of release in simulated colonic fluid. A fluorescence tracer indicated microspheres had high retention time of more than 20 h in the colon. Meanwhile, puerarin-loaded alginate microspheres not only significantly decreased the inflammatory response by downregulating the levels of pro-tumorigenic cytokines, but they reduced tumorigenesis and metastasis by inhibiting epithelial-mesenchymal transitions in AOM/DSS-induced colitis-associated colorectal cancer in mice. The overall results suggested that puerarin-loaded alginate microspheres could effectively inhibit development of colonic tumors, which could be developed as a promising therapeutic strategy for colitis-associated colorectal cancer.
Collapse
Affiliation(s)
- Xiao-Qing Deng
- Tianjin State Key Laboratory of Modern Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Hong-Bo Zhang
- Department of Medical Oncology, Tianjin Hospital of ITCWM, Nankai Hospital, Tianjin 300100, PR China
| | - Gui-Fang Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Dong Xu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Wen-Yan Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Qiang-Song Wang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, PR China.
| | - Yuan-Lu Cui
- Tianjin State Key Laboratory of Modern Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
11
|
Ramamoorthy P, Thomas SM, Kaushik G, Subramaniam D, Chastain KM, Dhar A, Tawfik O, Kasi A, Sun W, Ramalingam S, Gunewardena S, Umar S, Mammen JM, Padhye SB, Weir SJ, Jensen RA, Sittampalam GS, Anant S. Metastatic Tumor-in-a-Dish, a Novel Multicellular Organoid to Study Lung Colonization and Predict Therapeutic Response. Cancer Res 2019; 79:1681-1695. [PMID: 30674533 PMCID: PMC6445669 DOI: 10.1158/0008-5472.can-18-2602] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/11/2018] [Accepted: 01/17/2019] [Indexed: 12/22/2022]
Abstract
Metastasis is a major cause of cancer-related deaths. A dearth of preclinical models that recapitulate the metastatic microenvironment has impeded the development of therapeutic agents that are effective against metastatic disease. Because the majority of solid tumors metastasize to the lung, we developed a multicellular lung organoid that mimics the lung microenvironment with air sac-like structures and production of lung surfactant protein. We used these cultures, called primitive lung-in-a-dish (PLiD), to recreate metastatic disease using primary and established cancer cells. The metastatic tumor-in-a-dish (mTiD) cultures resemble the architecture of metastatic tumors in the lung, including angiogenesis. Pretreating PLiD with tumor exosomes enhanced cancer cell colonization. We next tested the response of primary and established cancer cells to current chemotherapeutic agents and an anti-VEGF antibody in mTiD against cancer cells in two-dimensional (2D) or 3D cultures. The response of primary patient-derived colon and ovarian tumor cells to therapy in mTiD cultures matched the response of the patient in the clinic, but not in 2D or single-cell-type 3D cultures. The sensitive mTiD cultures also produced significantly lower circulating markers for cancer similar to that seen in patients who responded to therapy. Thus, we have developed a novel method for lung colonization in vitro, a final stage in tumor metastasis. Moreover, the technique has significant utility in precision/personalized medicine, wherein this phenotypic screen can be coupled with current DNA pharmacogenetics to identify the ideal therapeutic agent, thereby increasing the probability of response to treatment while reducing unnecessary side effects. SIGNIFICANCE: A lung organoid that exhibits characteristics of a normal human lung is developed to study the biology of metastatic disease and therapeutic intervention.
Collapse
Affiliation(s)
- Prabhu Ramamoorthy
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
- Department of General Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Sufi Mary Thomas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas
| | - Gaurav Kaushik
- Department of General Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Dharmalingam Subramaniam
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
- Department of General Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Katherine M Chastain
- Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri
| | - Animesh Dhar
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Ossama Tawfik
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Anup Kasi
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Weijing Sun
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Satish Ramalingam
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Shahid Umar
- Department of General Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Joshua M Mammen
- Department of General Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Subhash B Padhye
- Interdisciplinary Science and Technology Research Academy, University of Pune, Pune, Maharashtra, India
| | - Scott J Weir
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Roy A Jensen
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - G Sitta Sittampalam
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland
| | - Shrikant Anant
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas.
- Department of General Surgery, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
12
|
Cui W, Li F, Yuan Q, Chen G, Chen C, Yu B. Role of VEGFA gene polymorphisms in colorectal cancer patients who treated with bevacizumab. Oncotarget 2017; 8:105472-105478. [PMID: 29285265 PMCID: PMC5739652 DOI: 10.18632/oncotarget.22295] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/26/2017] [Indexed: 01/05/2023] Open
Abstract
Objectives This study aimed to explore the effects of vascular endothelial growth factor A (VEGFA) gene polymorphisms (rs699947 and rs833061) on Bevacizumab (BEV) treatment in colorectal cancer (CRC) patients. Methods 125 CRC cases receiving BEV plus FOLFIRI treatment were recruited in this study. VEGFA polymorphisms were genotyped using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Correlation of VEGFA gene polymorphisms with the response rate and progression free survival (PFS) was evaluated. Multivariate analyses were performed to estimate the effects of VEGFA polymorphisms on the therapeutic effects of BEV treatment in CRC patients. Results Rs699947 variants did not show significant association with BEV treatment. For rs833061 analysis, TT and TC genotype carriers had significantly higher ORR (objective response rate) than CC carriers (P=0.048 and P=0.021, respectively). Moreover, TT carriers underwent a well DCR (disease control rate) compared to CC carriers (P=0.002). PFS time also showed obvious correlation with rs833061 polymorphism (log rank test, P=0.002). Multivariate analyses demonstrated that TT and TC genotypes of rs833061 polymorphism were significantly correlated with enhanced therapeutic effects and prolonged PFS in CRC patients. Conclusion VEGFA rs833061 polymorphism is significantly associated with the therapeutic efficiency of bevacizumab in CRC patients.
Collapse
Affiliation(s)
- Wei Cui
- Department of General Surgery, The Military General Hospital of Beijing PLA, Beijing 100700, China
| | - Feng Li
- Department of Health, The Military General Hospital of Beijing PLA, Beijing 100700, China
| | - Qiang Yuan
- Department of General Surgery, The Military General Hospital of Beijing PLA, Beijing 100700, China
| | - Gang Chen
- Department of General Surgery, The Military General Hospital of Beijing PLA, Beijing 100700, China
| | - Cailing Chen
- Department of General Surgery, The Military General Hospital of Beijing PLA, Beijing 100700, China
| | - Bo Yu
- Department of General Surgery, The Military General Hospital of Beijing PLA, Beijing 100700, China
| |
Collapse
|
13
|
Bahrami A, Hassanian SM, ShahidSales S, Farjami Z, Hasanzadeh M, Anvari K, Aledavood A, Maftouh M, Ferns GA, Khazaei M, Avan A. Targeting RAS signaling pathway as a potential therapeutic target in the treatment of colorectal cancer. J Cell Physiol 2017; 233:2058-2066. [PMID: 28262927 DOI: 10.1002/jcp.25890] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/02/2017] [Indexed: 12/19/2022]
Abstract
The V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) is frequently dysregulated in colorectal cancer (CRC). It is involved in the modulation of several downstream effectors, that include: Raf/Mek/Erk, PI3K/Akt, RalGDS/p38MAPK, and Rac/Rho, and thereby influences tumorigenesis, the invasive behaviors of tumor cell, and resistance to therapy. There is growing evidence exploring the use of drugs that target these pathways in the treatment of CRC. Cetuximab has been approved for CRC patients without a KRAS mutation, or for EGFR-expressing metastatic CRC, although some of the patients have a mutation of KRAS and NRAS. This review summarizes the recent knowledge about the therapeutic potential of targeting RAS with particular emphasis on recent preclinical and clinical studies in treatment of CRC.
Collapse
Affiliation(s)
- Afsane Bahrami
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodabeh ShahidSales
- Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Farjami
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Hasanzadeh
- Department of Gynecology Oncology, Woman Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kazem Anvari
- Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Aledavood
- Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftouh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, UK
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
14
|
Apsangikar P, Chaudhry S, Naik M, Deoghare S, Joseph J. Comparative pharmacokinetics, efficacy, and safety of bevacizumab biosimilar to reference bevacizumab in patients with metastatic colorectal cancer. Indian J Cancer 2017; 54:535-538. [DOI: 10.4103/ijc.ijc_394_17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
15
|
Olivieri M, Amata E, Vinciguerra S, Fiorito J, Giurdanella G, Drago F, Caporarello N, Prezzavento O, Arena E, Salerno L, Rescifina A, Lupo G, Anfuso CD, Marrazzo A. Antiangiogenic Effect of (±)-Haloperidol Metabolite II Valproate Ester [(±)-MRJF22] in Human Microvascular Retinal Endothelial Cells. J Med Chem 2016; 59:9960-9966. [PMID: 27739690 DOI: 10.1021/acs.jmedchem.6b01039] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
(±)-MRJF22 [(±)-2], a novel prodrug of haloperidol metabolite II (sigma-1 receptor antagonist/sigma-2 receptor agonist ligand) obtained by conjugation to valproic acid (histone deacetylase inhibitor) via an ester bond, exhibits antiangiogenic activity, being able to reduce human retinal endothelial cell (HREC) viability in a comparable manner to bevacizumab. Moreover, (±)-2 was able to significantly reduce viable cells count, endothelial cell migration, and tube formation in vascular endothelial growth factor A (VEGF-A) stimulated HREC cultures.
Collapse
Affiliation(s)
- Melania Olivieri
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Emanuele Amata
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Shila Vinciguerra
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Jole Fiorito
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Giovanni Giurdanella
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Nunzia Caporarello
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Emanuela Arena
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Loredana Salerno
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Antonio Rescifina
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Gabriella Lupo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Carmelina Daniela Anfuso
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Agostino Marrazzo
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|