1
|
Skaličková M, Abramenko N, Charnavets T, Vellieux F, Leischner Fialová J, Kučnirová K, Kejík Z, Masařík M, Martásek P, Pacak K, Pacák T, Jakubek M. Interaction of Selected Anthracycline and Tetracycline Chemotherapeutics with Poly(I:C) Molecules. ACS OMEGA 2025; 10:15935-15946. [PMID: 40321536 PMCID: PMC12044458 DOI: 10.1021/acsomega.4c05483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 02/11/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025]
Abstract
Despite the natural ability of the immune system to recognize cancer and, in some patients, even to eliminate it, cancer cells have acquired numerous evading mechanisms. With the increasing knowledge and focus shifting from targeting rapidly proliferating cells with chemotherapy to modulating the immune system, there have been recent efforts to integrate (e.g., simultaneously or sequentially) various therapeutic approaches. Combining the oncolytic activity of some chemotherapeutics with immunostimulatory molecules, so-called chemoimmunotherapy, is an attractive strategy. An example of such an immunostimulatory molecule is polyinosinic:polycytidylic acid [Poly(I:C)], a synthetic analogue of double-stranded RNA characterized by rapid nuclease degradation hampering its biological activity. This study investigated the possible interactions of tetracycline and anthracycline chemotherapeutics with different commercial Poly(I:C) molecules and protection against nuclease degradation. Fluorescence spectroscopy and circular dichroism revealed an interaction of all of the selected chemotherapeutics with Poly(I:C)s and the ability of doxycycline and minocycline to prolong the resistance to RNase cleavage, respectively. The partial protection was observed in vitro as well.
Collapse
Affiliation(s)
- Markéta Skaličková
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Nikita Abramenko
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Tatsiana Charnavets
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, 252
50 Vestec, Czech
Republic
| | - Frédéric Vellieux
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | | | - Kateřina Kučnirová
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Zdeněk Kejík
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Michal Masařík
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
- Department
of Physiology, Faculty of Medicine, Masaryk
University, Kamenice 5, 625 00 Brno, Czech Republic
- Department
of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625
00, Czech Republic
| | - Pavel Martásek
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Karel Pacak
- Section on
Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute
of Child Health and Human Development, National
Institutes of Health, Building 10, Room 1-3140, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Tomáš Pacák
- TumorSHOT, Italská 2581/67, Vinohrady,
Praha 2, Prague 120 00, Czech Republic
| | - Milan Jakubek
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| |
Collapse
|
2
|
Azarian M, Ramezani Farani M, C Cho W, Asgharzadeh F, Yang YJ, Moradi Binabaj M, M Tambuwala M, Farahani N, Hushmandi K, Huh YS. Advancements in colorectal cancer treatment: The role of metal-based and inorganic nanoparticles in modern therapeutic approaches. Pathol Res Pract 2024; 264:155706. [PMID: 39527908 DOI: 10.1016/j.prp.2024.155706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/17/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Recent advances in the treatment of colorectal cancer (CRC) have highlighted the integration of metal-based nanoparticles into sophisticated therapeutic strategies. This examination delves into the potential applications of these nanoparticles, particularly in augmenting the effectiveness of photodynamic therapy (PDT) and targeted drug delivery systems. Metal nanoparticles, such as gold (Au), silver (Ag), and copper (Cu), possess distinctive characteristics that make them valuable in cancer treatment. Beyond their role as drug carriers, these nanoparticles actively engage in therapeutic processes like apoptosis induction, enhancement of photothermal effects, and generation of reactive oxygen species (ROS) crucial for tumor cell eradication. The utilization of metal nanoparticles in CRC therapy addresses significant challenges encountered with conventional treatments, such as drug resistance and systemic toxicity. For example, engineered Au nanoparticles enable targeted drug delivery, reducing off-target effects and maximizing therapeutic efficacy against cancerous cells. Their capacity to absorb near-infrared light allows for localized hyperthermia, effectively eliminating cancerous tissues. Similarly, Cu nanoparticles exhibit potential in overcoming drug resistance by enhancing the efficacy of traditional chemotherapeutic agents through ROS production and improved drug stability. This review underscores the significance of precision medicine in CRC care. Through the integration of metal nanoparticles alongside complementary biomarkers and personalized treatment strategies, a more efficient and tailored therapeutic approach can be achieved. The synergistic effect of PDT in combination with metal nanoparticles introduces a novel methodology to CRC treatment, offering a dual-action mechanism that enhances tumor targeting while minimizing undesirable effects. In conclusion, the integration of metal-based nanoparticles in CRC therapy marks a significant progress in oncological treatments. Continued research is imperative to comprehensively grasp their mechanisms, optimize their clinical utility, and address potential safety considerations. This thorough assessment aims to pave the way for future advancements in CRC treatment through the application of nanotechnology and personalized medicine strategies.
Collapse
Affiliation(s)
- Maryam Azarian
- Department of Bioanalytical Ecotoxicology,UFZ- Helmholtz Centre for Environmental Research, Leipzig, Germany; Department of Radiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Fereshteh Asgharzadeh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yu-Jeong Yang
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Maryam Moradi Binabaj
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research Center, Gonabad University of Medical Science, Gonabad, Iran
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, United Kingdom
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
3
|
Zhang Z, Wang K, Liu M, Hu P, Xu Y, Yin D, Yang Y, Dong X, Qu C, Zhang L, Ni J, Yin X. Phototherapeutic effect of transformable peptides containing pheophorbide a on colorectal cancer. Drug Deliv 2022; 29:1608-1619. [PMID: 35612320 PMCID: PMC9135428 DOI: 10.1080/10717544.2022.2075987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Photodynamic therapy (PDT) and photothermal therapy (PTT) have attracted research interest for their noninvasive nature and selective treatment of tumor tissues. They are effective through the generation of reactive oxygen species (ROS) or heat. Nevertheless, several problems, including low bioavailability and long-lasting cutaneous photosensitivity, have limited their clinical application. In this study, we reported an in situ self-assembly strategy that could improve various biological properties of the photosensitizer in vivo. A photosensitizer connected to a receptor-mediated smart peptide can self-assemble into nanoparticles (NPs) under the force of hydrophobic interaction and then transform into a nanofibrillar network after attaching to the tumor cell surface with the help of the β-sheet-forming peptide KLVFF. The supramolecular structural changes deeply affected the PDT and PTT properties of the photosensitizer on tumors. After being aggregated into the nanostructure, the water solubility and targeting ability of the photosensitizer was ameliorated. Moreover, the improvement of the photothermal conversion efficiency, ROS generation, and tumor retention followed the formation of nanofibrils (NFs). This self-assembly strategy showed the ability of supramolecular nanofibrils to improve the bioavailability of photosensitizers, which provides a new potential treatment avenue for various cancer therapies.
Collapse
Affiliation(s)
- Zhiqin Zhang
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Kaixin Wang
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Manting Liu
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Panxiang Hu
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Yuchen Xu
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Dongge Yin
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Yuchang Yang
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoxv Dong
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Changhai Qu
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Lu Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Jian Ni
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing, China
| | - Xingbin Yin
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Tougeron D, Emambux S, Favot L, Lecomte T, Wierzbicka-Hainaut E, Samimi M, Frouin E, Azzopardi N, Chevrier J, Serres L, Godet J, Levillain P, Paintaud G, Ferru A, Rouleau L, Delwail A, Silvain C, Tasu JP, Morel F, Ragot S, Lecron JC. Skin inflammatory response and efficacy of anti-epidermal growth factor receptor therapy in metastatic colorectal cancer (CUTACETUX). Oncoimmunology 2020; 9:1848058. [PMID: 33299659 PMCID: PMC7714491 DOI: 10.1080/2162402x.2020.1848058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/17/2020] [Accepted: 11/06/2020] [Indexed: 12/22/2022] Open
Abstract
Anti-epidermal growth factor receptor (EGFR) monoclonal antibody is a standard treatment of metastatic colorectal cancer (mCRC) and its most common adverse effect is a papulopustular acneiform rash. The aim of the CUTACETUX study was to characterize the skin inflammatory response associated with this rash and its relation to treatment efficacy. This prospective study included patients with mCRC treated with first-line chemotherapy plus cetuximab. Patients underwent skin biopsies before the initiation of cetuximab (D0) and before the third infusion (D28), one in a rash zone and one in an unaffected zone. Expression of Th17-related cytokines (IL-17A, IL-21, IL-22), antimicrobial peptides (S100A7 and BD-2), innate response-related cytokines (IL-1β, IL-6, TNF-α and OSM), T-reg-related cytokines (IL-10 and TGF-β), Th1-related cytokine (IFN-γ), Th2-related cytokine (IL-4), Thymic stromal lymphopoietin and keratinocyte-derived cytokines (IL-8, IL-23 and CCL20) were determined by RT-PCR. Twenty-seven patients were included. Levels of most of the cytokines increased at D28 in the rash zone compared to D0. No significant association was observed between variations of cytokines levels and treatment response in the rash zone and only the increase of IL-4 (p = .04) and IL-23 (p = .02) levels between D0 and D28 in the unaffected zone was significantly associated with treatment response. Increased levels of IL-8 (p = .02), BD-2 (p = .02), IL-1β (p = .004) and OSM (p = .02) in the rash zone were associated with longer progression-free survival. Expression of Th2-related and keratinocyte-derived cytokines in the skin was associated with anti-EGFR efficacy. If this inflammatory signature can explain the rash, the exact mechanism by which these cytokines are involved in anti-EGFR tumor response remains to be studied.
Collapse
Affiliation(s)
- David Tougeron
- Gastroenterology Department, Poitiers University Hospital, Poitiers, France
- Laboratory Inflammation, Tissus Epithéliaux Et Cytokines, EA 4331, Poitiers University, Poitiers, France
| | - Sheik Emambux
- Medical Oncology Department, Poitiers University Hospital, Poitiers, France
| | - Laure Favot
- Laboratory Inflammation, Tissus Epithéliaux Et Cytokines, EA 4331, Poitiers University, Poitiers, France
| | - Thierry Lecomte
- Gastroenterology Department, Tours University Hospital, Tours, France
- Tours University, EA7501 GICC, Team PATCH, Tours, France, Tours, France
| | - Ewa Wierzbicka-Hainaut
- Laboratory Inflammation, Tissus Epithéliaux Et Cytokines, EA 4331, Poitiers University, Poitiers, France
- Dermatology Department, Poitiers University Hospital, Poitiers, France
| | - Mahtab Samimi
- Dermatology Department, Tours University Hospital, ISP1282 INRA, Université De Tours, Tours, France
| | - Eric Frouin
- Laboratory Inflammation, Tissus Epithéliaux Et Cytokines, EA 4331, Poitiers University, Poitiers, France
- Pathology Department, Poitiers University Hospital, Poitiers, France
| | - Nicolas Azzopardi
- Tours University, EA7501 GICC, Team PATCH, Tours, France, Tours, France
| | - Jocelyn Chevrier
- Laboratory Inflammation, Tissus Epithéliaux Et Cytokines, EA 4331, Poitiers University, Poitiers, France
| | - Laura Serres
- Gastroenterology Department, Poitiers University Hospital, Poitiers, France
| | - Julie Godet
- Pathology Department, Poitiers University Hospital, Poitiers, France
| | - Pierre Levillain
- Pathology Department, Poitiers University Hospital, Poitiers, France
| | - Gilles Paintaud
- Tours University, EA7501 GICC, Team PATCH, Tours, France, Tours, France
- Pharmacology Department, Tours University Hospital, Tours, France
| | - Aurélie Ferru
- Medical Oncology Department, Poitiers University Hospital, Poitiers, France
| | - Laetitia Rouleau
- Gastroenterology Department, Poitiers University Hospital, Poitiers, France
| | - Adriana Delwail
- ImageUP, Plate-forme d’Imagerie and Laboratoire Signalisation Et Transport Ioniques Membranaires ERL CNRS 7003/EA 7349, Poitiers University, Poitiers, France
| | - Christine Silvain
- Gastroenterology Department, Poitiers University Hospital, Poitiers, France
- Laboratory Inflammation, Tissus Epithéliaux Et Cytokines, EA 4331, Poitiers University, Poitiers, France
| | - Jean-Pierre Tasu
- Radiology Department, Poitiers University Hospital, Poitiers, France
- LaTIM, INSERM U1101, Brest, France
| | - Franck Morel
- Laboratory Inflammation, Tissus Epithéliaux Et Cytokines, EA 4331, Poitiers University, Poitiers, France
| | - Stéphanie Ragot
- Cic Inserm 1402, Poitiers University Hospital, Poitiers, France
| | - Jean-Claude Lecron
- Laboratory Inflammation, Tissus Epithéliaux Et Cytokines, EA 4331, Poitiers University, Poitiers, France
- Immunology/inflammation Laboratory, Poitiers University Hospital, Poitiers, France
| |
Collapse
|
5
|
Nardone V, Pastina P, Giannicola R, Agostino R, Croci S, Tini P, Pirtoli L, Giordano A, Tagliaferri P, Correale P. How to Increase the Efficacy of Immunotherapy in NSCLC and HNSCC: Role of Radiation Therapy, Chemotherapy, and Other Strategies. Front Immunol 2018; 9:2941. [PMID: 30619301 PMCID: PMC6299115 DOI: 10.3389/fimmu.2018.02941] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/30/2018] [Indexed: 12/14/2022] Open
Affiliation(s)
- Valerio Nardone
- Radiation Oncology Unit, University Hospital of Siena, Siena, Italy
| | | | - Rocco Giannicola
- Medical Oncology Unit, Grand Metropolitan Hospital "Bianchi Melacrino Morelli", Reggio Calabria, Italy
| | - Rita Agostino
- Medical Oncology Unit, Grand Metropolitan Hospital "Bianchi Melacrino Morelli", Reggio Calabria, Italy
| | - Stefania Croci
- Radiation Oncology Unit, University Hospital of Siena, Siena, Italy
| | - Paolo Tini
- Radiation Oncology Unit, University Hospital of Siena, Siena, Italy.,Sbarro Health Research Organization, Temple University, Philadelphia, PA, United States
| | - Luigi Pirtoli
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Antonio Giordano
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States.,Department of Medicine, Surgery and Neurosciences University of Siena, Siena, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.,Medical Oncology Unit, Azienda Ospedaliero - Universitaria "Mater Domini", Catanzaro, Italy
| | - Pierpaolo Correale
- Medical Oncology Unit, Grand Metropolitan Hospital "Bianchi Melacrino Morelli", Reggio Calabria, Italy
| |
Collapse
|
6
|
Gu R, He Y, Han S, Yuan S, An Y, Meng Z, Zhu X, Gan H, Wu Z, Li J, Zheng Y, Zhang L, Gao L, Dou G. Pharmacokinetics and bioavailability of tuftsin-derived T peptide, a promising antitumor agent, in beagles. Drug Metab Pharmacokinet 2015; 31:51-56. [PMID: 26775850 DOI: 10.1016/j.dmpk.2015.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/27/2015] [Accepted: 08/16/2015] [Indexed: 12/09/2022]
Abstract
Tuftsin, a natural phagocytosis-stimulating tetrapeptide, had aroused much interest in tumor immunotherapy, but the poor pharmacokinetics hampered its clinical developments, for that it was extremely susceptible to degradation by enzymolysis in vivo. T Peptide (TP) was a newly designed tuftsin derivative aimed to enhance stability and was proved to have significant antitumor activity. In this study, the pharmacokinetics and bioavailability of TP was first clarified in beagles with subcutaneous administration, by using a simple and robust competitive ELISA method. Dose-dependency and non-linear dynamics of TP after single-dose (2, 6 and 18 mg kg(-1), respectively) were found, and the half-time of TP was proved to reach 1.3-2.8 h. Multiple dosing of 6 mg kg(-1) once a day for 7 days resulted in a slight accumulation (accumulation index was 1.92 ± 0.43), indicating that the dosing interval in the following clinical trial needs to be extended. The absolute bioavailability of TP was 31.1 ± 6.2% after subcutaneous administration. These results first demonstrated the pharmacokinetics and bioavailability data of TP in vivo, which illustrated the potential druggability of TP and provided useful information for the dosage regimen design in the following clinical trials, as well as a simple and feasible analytical method for clinical sample analysis.
Collapse
Affiliation(s)
- Ruolan Gu
- State Key Laboratory of Drug Metabolism, Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Beijing 100850, PR China
| | - Yanlin He
- State Key Laboratory of Drug Metabolism, Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Beijing 100850, PR China
| | - Su Han
- College of Pharmacy, Nankai University, 94, Weijin Road, Tianjin 300071, PR China
| | - Shoujun Yuan
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, 27, Taiping Road, Beijing 100850, PR China
| | - Yinghong An
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, 27, Taiping Road, Beijing 100850, PR China; Center for Clinical Laboratory, Airforce General Hospital of Chinese PLA, 30, Fucheng Road, Beijing 100142, PR China
| | - Zhiyun Meng
- State Key Laboratory of Drug Metabolism, Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Beijing 100850, PR China
| | - Xiaoxia Zhu
- State Key Laboratory of Drug Metabolism, Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Beijing 100850, PR China
| | - Hui Gan
- State Key Laboratory of Drug Metabolism, Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Beijing 100850, PR China
| | - Zhuona Wu
- State Key Laboratory of Drug Metabolism, Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Beijing 100850, PR China
| | - Jian Li
- State Key Laboratory of Drug Metabolism, Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Beijing 100850, PR China
| | - Ying Zheng
- State Key Laboratory of Drug Metabolism, Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Beijing 100850, PR China
| | - Ling Zhang
- State Key Laboratory of Drug Metabolism, Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Beijing 100850, PR China
| | - Lei Gao
- State Key Laboratory of Drug Metabolism, Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Beijing 100850, PR China
| | - Guifang Dou
- State Key Laboratory of Drug Metabolism, Hematological Pharmacology, Beijing Institute of Transfusion Medicine, 27, Taiping Road, Beijing 100850, PR China.
| |
Collapse
|
7
|
Sakai H, Kokura S, Ishikawa T, Tsuchiya R, Okajima M, Matsuyama T, Adachi S, Katada K, Kamada K, Uchiyama K, Handa O, Takagi T, Yagi N, Naito Y, Yoshikawa T. Effects of anticancer agents on cell viability, proliferative activity and cytokine production of peripheral blood mononuclear cells. J Clin Biochem Nutr 2012; 52:64-71. [PMID: 23341700 PMCID: PMC3541421 DOI: 10.3164/jcbn.12-60] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 08/06/2012] [Indexed: 11/22/2022] Open
Abstract
We investigated the effects of anticancer agents on peripheral blood mononuclear cells for the purpose of providing data to support new translational chemoimmunotherapy regimens. Peripheral-blood mononuclear cells were treated with one of four anticancer agents (5-fluorouracil, irinotecan, cisplatin, and gemcitabine) for 2 h, after which cell viability was determined. For assessment of effects of each drug on proliferation and cytokine production, cells were stimulated with phytohemagglutinin for 48 h. As a result, the anticancer agents did not affect cell viability. Cell proliferation was unaffected by 5-fluorouracil and irinotecan but inhibited by cisplatin and gemcitabine. Treatment with gemcitabine enhanced the production of IFN-γ and decreased the number of regulatory T cells. gemcitabine treatment increased IFN-γ production among CD4 T cells but not among CD8 T cells. The results indicated that GEM had immunoregulatory properties that might support immune response against cancer. This finding has implications for designing chemoimmunotherapy strategies.
Collapse
Affiliation(s)
- Hiromi Sakai
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Kitamura H, Tsukamoto T. Immunotherapy for urothelial carcinoma: current status and perspectives. Cancers (Basel) 2011; 3:3055-72. [PMID: 24212945 PMCID: PMC3759186 DOI: 10.3390/cancers3033055] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 07/22/2011] [Accepted: 07/26/2011] [Indexed: 12/14/2022] Open
Abstract
Intravesical instillation of bacillus Calmette Guérin (BCG) for the treatment of urothelial carcinoma (UC) of the bladder is based on the BCG-induced immune response, which eradicates and prevents bladder cancer. The results of recent studies have suggested that not only major histocompatibility complex (MHC)-nonrestricted immune cells such as natural killer cells, macrophages, neutrophils, etc., but also MHC-restricted CD8+ T cells play an important role and are one of the main effectors in this therapy. Better understanding of the mechanism of BCG immunotherapy supports the idea that active immunotherapy through its augmented T cell response can have great potential for the treatment of advanced UC. In this review, progress in immunotherapy for UC is discussed based on data from basic, translational and clinical studies. We also review the escape mechanism of cancer cells from the immune system, and down-regulation of MHC class I molecules.
Collapse
Affiliation(s)
- Hiroshi Kitamura
- Department of Urology, Sapporo Medical University School of Medicine, South 1 West 16, Chuo-ku, Sapporo 060-8543, Japan.
| | | |
Collapse
|
9
|
Deschoolmeester V, Baay M, Lardon F, Pauwels P, Peeters M. Immune Cells in Colorectal Cancer: Prognostic Relevance and Role of MSI. CANCER MICROENVIRONMENT 2011; 4:377-92. [PMID: 21618031 DOI: 10.1007/s12307-011-0068-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 05/19/2011] [Indexed: 12/14/2022]
Abstract
There is growing evidence that both local and systemic inflammatory responses play an important role in the progression of a variety of solid tumors. Colorectal cancer (CRC) results from the cumulative effect of sequential genetic alterations, leading to the expression of tumor-associated antigens possibly inducing a cellular anti-tumor immune response. It is well recognized that cytotoxic lymphocytes (CTLs) constitute one of the most important effector mechanisms of anti-tumor-immunity. However, their potential prognostic influence in CRC remains controversial. In addition, other key players like natural killer cells, tumor associated macrophages and regulatory T cells play an important role in the immune attack against CRC and need further investigation. This review will mainly focus on the role of the adaptive immune system in CRC and particularly in regard to microsatellite instability.
Collapse
Affiliation(s)
- Vanessa Deschoolmeester
- Laboratory of Cancer Research and Clinical Oncology, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium,
| | | | | | | | | |
Collapse
|
10
|
Deschoolmeester V, Baay M, Specenier P, Lardon F, Vermorken JB. A review of the most promising biomarkers in colorectal cancer: one step closer to targeted therapy. Oncologist 2010; 15:699-731. [PMID: 20584808 PMCID: PMC3228001 DOI: 10.1634/theoncologist.2010-0025] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 05/01/2010] [Indexed: 02/06/2023] Open
Abstract
Rapidly growing insights into the molecular biology of colorectal cancer (CRC) and recent developments in gene sequencing and molecular diagnostics have led to high expectations for the identification of molecular markers to be used in optimized and tailored treatment regimens. However, many of the published data on molecular biomarkers are contradictory in their findings and the current reality is that no molecular marker, other than the KRAS gene in the case of epidermal growth factor receptor (EGFR)- targeted therapy for metastatic disease, has made it into clinical practice. Many markers investigated suffer from technical shortcomings, resulting from lack of quantitative techniques to capture the impact of the molecular alteration. This understanding has recently led to the more comprehensive approaches of global gene expression profiling or genome-wide analysis to determine prognostic and predictive signatures in tumors. In this review, an update of the most recent data on promising biological prognostic and/or predictive markers, including microsatellite instability, epidermal growth factor receptor, KRAS, BRAF, CpG island methylator phenotype, cytotoxic T lymphocytes, forkhead box P3-positive T cells, receptor for hyaluronic acid-mediated motility, phosphatase and tensin homolog, and T-cell originated protein kinase, in patients with CRC is provided.
Collapse
Affiliation(s)
- Vanessa Deschoolmeester
- Laboratory of Cancer Research and Clinical Oncology, Department of Medical Oncology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| | | | | | | | | |
Collapse
|
11
|
Correale P, Tagliaferri P, Fioravanti A, Del Vecchio MT, Remondo C, Montagnani F, Rotundo MS, Ginanneschi C, Martellucci I, Francini E, Cusi MG, Tassone P, Francini G. Immunity feedback and clinical outcome in colon cancer patients undergoing chemoimmunotherapy with gemcitabine + FOLFOX followed by subcutaneous granulocyte macrophage colony-stimulating factor and aldesleukin (GOLFIG-1 Trial). Clin Cancer Res 2008; 14:4192-9. [PMID: 18593999 DOI: 10.1158/1078-0432.ccr-07-5278] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE GOLFIG chemoimmunotherapy regimen proved to be a safe and very active chemoimmunotherapy regimen in advanced colon cancer patients. We have thus investigated the immunobiological feedback to the treatment and its possible correlation with the clinical outcome of these patients. EXPERIMENTAL DESIGN This clinical and immunologic study involved 46 patients, 27 males and 19 females, enrolled in the GOLFIG-1 phase II trial who received gemcitabine (1,000 mg/m(2) on days 1 and 15), oxaliplatin (85 mg/m(2) on days 2 and 16), levofolinic acid (100 mg/m(2) on days 1, 2, 15, and 16), and 5-fluorouracil (400 mg/m(2) as a bolus, and 800 mg/m(2) as a 24-hour infusion on days 1, 2, 15, and 16) followed by s.c. granulocyte macrophage colony-stimulating factor (100 mug, on days 3-7) and interleukin 2 (0.5 x 10(6) IU twice a day on days 8-14 and 17-29). RESULTS The regimen was confirmed to be safe and very active in pretreated patients with metastatic colorectal cancer. A subgroup analysis of these patients revealed a prolonged time to progression and survival in six patients who developed late signs of autoimmunity. A multivariate analysis validated the occurrence of autoimmunity signs as an independent predictor of favorable outcome. A parallel immunologic study detected in the peripheral blood mononuclear cells of these patients a progressive increase in lymphocyte and eosinophil counts, amplification in central memory, a marked depletion of immunosuppressive regulatory T cells, and activation of colon cancer-specific cytotoxic T cells. CONCLUSIONS Our results suggest that immunity feedback to GOLFIG regimen and its antitumor activity are tightly correlated.
Collapse
Affiliation(s)
- Pierpaolo Correale
- Section of Medical Oncology, Department "Giorgio Segre" of Pharmacology, Rheumatology Unit, Siena University School of Medicine, Siena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Correale P, Fioravanti A, Bertoldi I, Montagnani F, Miracco C, Francini G. Occurrence of autoimmunity in a long-term survivor with metastatic colon carcinoma treated with a new chemo-immunotherapy regimen. J Chemother 2008; 20:278-81. [PMID: 18467257 DOI: 10.1179/joc.2008.20.2.278] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
GOLFIG-1 chemo-immunotherapy is a new translational anticancer regimen based on the combined use of gemcitabine, oxalipatin, levofolinic acid and infusional 5-fluorouracil together with the subcutaneous administration immunoadjuvant cytokines (GM-CSF and ultra low dose IL-2). This regimen, tested in a phase II trial, was safe and very active in patients with metastatic colorectal carcinoma and it has been shown to have powerful immunobiological activity. Treatment with the GOLFIG regimen resulted in the induction of a colon cancer specific cell mediated immune response associated with a significant reduction in the percentage of peripheral regulatory T (T(reg)) cells, a very immunosuppressive lymphocyte subset which is commonly over-represented in cancer patients. These cells are able to prevent the occurrence of autoimmunity in response to immunological stimuli, thus their malfunctioning has been associated with the occurrence of auto-immune diseases but may also be responsible for more efficient anticancer immune reaction. In this manuscript we describe a clinical case concerning a patient with metastatic colon carcinoma who responded to the GOLFIG regimen, showed symptoms of autoimmunity [Discoid Lupus Erythematosus (DLE)] and had a very long survival.
Collapse
Affiliation(s)
- P Correale
- Section of Medical Oncology, Department of Clinical Medicine and Immunological Sciences, Siena University School of Medicine, Siena, Italy
| | | | | | | | | | | |
Collapse
|