1
|
Giram PS, Nimma R, Bulbule A, Yadav AS, Gorain M, Venkata Radharani NN, Kundu GC, Garnaik B. Engineered PLGA Core-Lipid Shell Hybrid Nanocarriers Improve the Efficacy and Safety of Irinotecan to Combat Colon Cancer. ACS Biomater Sci Eng 2024; 10:6661-6676. [PMID: 39269431 DOI: 10.1021/acsbiomaterials.4c01260] [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] [Indexed: 09/15/2024]
Abstract
Poly(lactide-co-glycolide) (PLGA) is a biocompatible and biodegradable copolymer that has gained high acceptance in biomedical applications. In the present study, PLGA (Mw = 13,900) was synthesized by ring-opening polymerization in the presence of a biocompatible zinc-proline initiator through a green route. Irinotecan (Ir) loaded with efficient PLGA core-lipid shell hybrid nanocarriers (lipomers, LPs) were formulated with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino (polyethylene glycol)-2000] (DSPE-PEG-2000), using soya lecithin, by a nanoprecipitation method, and the fabricated LPs were designated as P-DSPE-Ir and P-DSPE-PEG-Ir, respectively. The formulated LPs were further validated for their physicochemical properties and biological potential for colon cancer application. The potential delivery of a poorly water-soluble chemotherapeutic drug (Ir) was studied for the treatment of colon cancer. LPs were successfully prepared, providing controlled size (80-120 nm) and surface charge (∼ -35 mV), and the sustained release properties and cytotoxicity against CT-26 colon cancer cells were studied. The in vivo biodistribution and tumor site retention in CT-26 xenograft tumor-bearing Balb/C mice showed promising results for tumor uptake and retention for a prolonged time period. Unlike P-DSPE-Ir, the P-DSPE-PEG-Ir LP exhibited significant tumor growth delay as compared to untreated and blank formulation-treated groups in CT-26 (subcutaneous tumor model) after 4 treatments of 10 mg irinotecan/kg dose. The biocompatibility and safety of the LPs were confirmed by an acute toxicity study of the optimized formulation. Overall, this proof-of-concept study demonstrates that the PLGA-based LPs improve the efficacy and bioavailability and decrease neutropenia of Ir to combat colon cancer.
Collapse
Affiliation(s)
- Prabhanjan S Giram
- Polymer Science and Engineering Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research AcSIR Headquarters, CSIR-HRDC Campus Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| | - Ramakrishna Nimma
- Laboratory of Tumor, Biology, Angiogenesis and Nanomedicine Research, National Center for Cell Science, Pune 411007, India
| | - Anuradha Bulbule
- Laboratory of Tumor, Biology, Angiogenesis and Nanomedicine Research, National Center for Cell Science, Pune 411007, India
| | - Amit Singh Yadav
- Laboratory of Tumor, Biology, Angiogenesis and Nanomedicine Research, National Center for Cell Science, Pune 411007, India
| | - Mahadeo Gorain
- Laboratory of Tumor, Biology, Angiogenesis and Nanomedicine Research, National Center for Cell Science, Pune 411007, India
| | | | - Gopal C Kundu
- School of Biotechnology and Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to be University, Institute of Eminence, Bhubaneswar 751 024, India
| | - Baijayantimala Garnaik
- Polymer Science and Engineering Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research AcSIR Headquarters, CSIR-HRDC Campus Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| |
Collapse
|
2
|
Di Dalmazi G, Giuliani C, Bucci I, Mascitti M, Napolitano G. Promising Role of Alkaloids in the Prevention and Treatment of Thyroid Cancer and Autoimmune Thyroid Disease: A Comprehensive Review of the Current Evidence. Int J Mol Sci 2024; 25:5395. [PMID: 38791433 PMCID: PMC11121374 DOI: 10.3390/ijms25105395] [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: 03/21/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Thyroid cancer (TC) and thyroid autoimmune disorders (AITD) are among the most common diseases in the general population, with higher incidence in women. Chronic inflammation and autoimmunity play a pivotal role in carcinogenesis. Some studies, indeed, have pointed out the presence of AITD as a risk factor for TC, although this issue remains controversial. Prevention of autoimmune disease and cancer is the ultimate goal for clinicians and scientists, but it is not always feasible. Thus, new treatments, that overcome the current barriers to prevention and treatment of TC and AITD are needed. Alkaloids are secondary plant metabolites endowed with several biological activities including anticancer and immunomodulatory properties. In this perspective, alkaloids may represent a promising source of prophylactic and therapeutic agents for TC and AITD. This review encompasses the current published literature on alkaloids effects on TC and AITD, with a specific focus on the pathways involved in TC and AITD development and progression.
Collapse
Affiliation(s)
- Giulia Di Dalmazi
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Cesidio Giuliani
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Ines Bucci
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Marco Mascitti
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Giorgio Napolitano
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
3
|
Zhang P, Tao C, Shimura T, Huang AC, Kong N, Dai Y, Yao S, Xi Y, Wang X, Fang J, Moses MA, Guo P. ICAM1 antibody drug conjugates exert potent antitumor activity in papillary and anaplastic thyroid carcinoma. iScience 2023; 26:107272. [PMID: 37520726 PMCID: PMC10371847 DOI: 10.1016/j.isci.2023.107272] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/27/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
Treatment options for anaplastic thyroid cancer (ATC) and refractory papillary thyroid carcinoma (PTC) are limited and outcomes remain poor. In this study, we determined via bioinformatic expression analyses and immunohistochemistry staining that intercellular adhesion molecule-1(ICAM1) is an attractive target for ATC and PTC. We designed and engineered two ICAM1-directed antibody-drug conjugate (I1-MMAE and I1-DXd), both of which potently and selectively ablate multiple human ATC and PTC cell lines without affecting non-plastic cells in vitro. Furthermore, I1-MMAE and I1-DXd mediated a potent tumor regression in ATC and PTC xenograft models. To develop a precision medicine, we also explored magnetic resonance imaging (MRI) as a non-invasive biomarker detection method to quantitatively map ICAM1 antigen expression in heterogeneous thyroid tumors. Taken together, this study provides a strong rationale for the further development of I1-MMAE and I1-DXd as promising therapeutic candidates to treat advanced PTC and ATC.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Medical Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Changjuan Tao
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Department of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | | | - Nana Kong
- MabPlex International, Yantai, Shandong 264006, China
| | - Yujie Dai
- MabPlex International, Yantai, Shandong 264006, China
| | - Shili Yao
- School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Yun Xi
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Xing Wang
- Department of Head and Neck Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Marsha A. Moses
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Peng Guo
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
4
|
Mendes I, Vale N. How Can the Microbiome Induce Carcinogenesis and Modulate Drug Resistance in Cancer Therapy? Int J Mol Sci 2023; 24:11855. [PMID: 37511612 PMCID: PMC10380870 DOI: 10.3390/ijms241411855] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Over the years, cancer has been affecting the lives of many people globally and it has become one of the most studied diseases. Despite the efforts to understand the cell mechanisms behind this complex disease, not every patient seems to respond to targeted therapies or immunotherapies. Drug resistance in cancer is one of the limiting factors contributing to unsuccessful therapies; therefore, understanding how cancer cells acquire this resistance is essential to help cure individuals affected by cancer. Recently, the altered microbiome was observed to be an important hallmark of cancer and therefore it represents a promising topic of cancer research. Our review aims to provide a global perspective of some cancer hallmarks, for instance how genetic and epigenetic modifications may be caused by an altered human microbiome. We also provide information on how an altered human microbiome can lead to cancer development as well as how the microbiome can influence drug resistance and ultimately targeted therapies. This may be useful to develop alternatives for cancer treatment, i.e., future personalized medicine that can help in cases where traditional cancer treatment is unsuccessful.
Collapse
Affiliation(s)
- Inês Mendes
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- School of Life and Environmental Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Edifício de Geociências, 5000-801 Vila Real, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
5
|
Lee YM, Chen YH, Ou DL, Hsu CL, Liu JH, Ko JY, Hu MCT, Tan CT. SN-38, an active metabolite of irinotecan, enhances anti-PD-1 treatment efficacy in head and neck squamous cell carcinoma. J Pathol 2023; 259:428-440. [PMID: 36641765 DOI: 10.1002/path.6055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 12/08/2022] [Accepted: 01/12/2023] [Indexed: 01/16/2023]
Abstract
Anti-programmed cell death 1 (anti-PD-1) therapy shows definite but modest activity in patients with advanced/metastatic head and neck squamous cell carcinoma (HNSCC). Preliminary evidence suggests that SN-38, an activated form of irinotecan that increases expression of the transcription factor FoxO3a, can suppress programmed cell death ligand-1 (PD-L1) expression in breast and ovarian tumor models. We analyzed the SN-38-mediated activation of natural killer cells in vitro and explored the efficacy of SN-38 in combination with anti-PD-1 for treatment in vivo. In vitro, SN-38 enhanced the expression of FoxO3a and reduced the expression of c-Myc and PD-L1 dose-dependently in tumor cells. Low-dose SN-38 increased interferon-γ secretion by NK cells and promoted NK cell-mediated cytotoxicity in tumor cells. In vivo studies revealed that at non-cytotoxic drug concentrations, SN-38 significantly enhanced anti-PD-1 activity in suppressing murine tumor growth. We found increased NK cell and CD8+ T-cell infiltration in post-treatment tumors. RNA-seq analysis indicated that SN-38 increased the enrichment of immune cells and biological function genes related to the immune responses. SN-38 is a potentially beneficial adjunct to checkpoint inhibitor therapy in HNSCC. Further studies exploring its mechanism of action and possible applications are necessary. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Yi-Mei Lee
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan.,Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Hsin Chen
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan.,Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Da-Liang Ou
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.,YongLin Institute of Health, National Taiwan University, Taipei, Taiwan
| | - Chia-Lang Hsu
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Jia-Hua Liu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan.,Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei, Taiwan
| | - Jenq-Yuh Ko
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Mickey C-T Hu
- Panorama Institute of Molecular Medicine, Sunnyvale, CA, USA.,Division of Gynecologic Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ching-Ting Tan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan.,Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei, Taiwan.,Department of Otolaryngology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Otolaryngology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| |
Collapse
|
6
|
Rationale Efficacy and Safety Evidence of Lenvatinib and Pembrolizumab Association in Anaplastic Thyroid Carcinoma. Curr Oncol 2022; 29:7718-7731. [PMID: 36290887 PMCID: PMC9601195 DOI: 10.3390/curroncol29100610] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 01/13/2023] Open
Abstract
Anaplastic thyroid carcinoma (ATC) are highly aggressive malignant tumors with poor overall prognosis despite multimodal therapy. As ATC are extremely rare, no randomized controlled study has been published for metastatic disease. Thyrosine kinase inhibitors, especially lenvatinib and immune checkpoint inhibitors such as pembrolizumab, are emerging drugs for ATC. Few studies have reported the efficacity of pembrolizumab and lenvatinib association, resulting in its frequent off-label use. In this review, we discuss rationale efficacy and safety evidence for the association of lenvatinib and pembrolizumab in ATC. First, we discuss preclinical rationale for pembrolizumab monotherapy, lenvatinib monotherapy and synergistic action of pembrolizumab and lenvatinib in the metastatic setting. We also discuss clinical evidence for immunotherapy and pembrolizumab in ATC through the analysis of studies evaluating immunotherapy, lenvatinib and pembrolizumab lenvatinib association in ATC. In addition, we discuss the safety of this association and potential predictive biomarkers of efficiency.
Collapse
|
7
|
Kryczka J, Boncela J. Integrated Bioinformatics Analysis of the Hub Genes Involved in Irinotecan Resistance in Colorectal Cancer. Biomedicines 2022; 10:biomedicines10071720. [PMID: 35885025 PMCID: PMC9312838 DOI: 10.3390/biomedicines10071720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/08/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
Different drug combinations including irinotecan remain some of the most important therapeutic modalities in treating colorectal cancer (CRC). However, chemotherapy often leads to the acquisition of cancer drug resistance. To bridge the gap between in vitro and in vivo models, we compared the mRNA expression profiles of CRC cell lines (HT29, HTC116, and LoVo and their respective irinotecan-resistant variants) with patient samples to select new candidate genes for the validation of irinotecan resistance. Data were downloaded from the Gene Expression Omnibus (GEO) (GSE42387, GSE62080, and GSE18105) and the Human Protein Atlas databases and were subjected to an integrated bioinformatics analysis. The protein–protein interaction (PPI) network of differently expressed genes (DEGs) between FOLFIRI-resistant and -sensitive CRC patients delivered several potential irinotecan resistance markers: NDUFA2, SDHD, LSM5, DCAF4, COX10 RBM8A, TIMP1, QKI, TGOLN2, and PTGS2. The chosen DEGs were used to validate irinotecan-resistant cell line models, proving their substantial phylogenetic heterogeneity. These results indicated that in vitro models are highly limited and favor different mechanisms than in vivo, patient-derived ones. Thus, cell lines can be perfectly utilized to analyze specific mechanisms on their molecular levels but cannot mirror the complicated drug resistance network observed in patients.
Collapse
|
8
|
Yue B, Gao R, Wang Z, Dou W. Microbiota-Host-Irinotecan Axis: A New Insight Toward Irinotecan Chemotherapy. Front Cell Infect Microbiol 2021; 11:710945. [PMID: 34722328 PMCID: PMC8553258 DOI: 10.3389/fcimb.2021.710945] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/23/2021] [Indexed: 12/19/2022] Open
Abstract
Irinotecan (CPT11) and its active metabolite ethyl-10-hydroxy-camptothecin (SN38) are broad-spectrum cytotoxic anticancer agents. Both cause cell death in rapidly dividing cells (e.g., cancer cells, epithelial cells, hematopoietic cells) and commensal bacteria. Therefore, CPT11 can induce a series of toxic side-effects, of which the most conspicuous is gastrointestinal toxicity (nausea, vomiting, diarrhea). Studies have shown that the gut microbiota modulates the host response to chemotherapeutic drugs. Targeting the gut microbiota influences the efficacy and toxicity of CPT11 chemotherapy through three key mechanisms: microbial ecocline, catalysis of microbial enzymes, and immunoregulation. This review summarizes and explores how the gut microbiota participates in CPT11 metabolism and mediates host immune dynamics to affect the toxicity and efficacy of CPT11 chemotherapy, thus introducing a new concept that is called "microbiota-host-irinotecan axis". Also, we emphasize the utilization of bacterial β-glucuronidase-specific inhibitor, dietary interventions, probiotics and strain-engineered interventions as emergent microbiota-targeting strategies for the purpose of improving CPT11 chemotherapy efficiency and alleviating toxicity.
Collapse
Affiliation(s)
- Bei Yue
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai, China
| | - Ruiyang Gao
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai, China
| | - Zhengtao Wang
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai, China
| | - Wei Dou
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai, China
| |
Collapse
|
9
|
De Leo S, Trevisan M, Fugazzola L. Recent advances in the management of anaplastic thyroid cancer. Thyroid Res 2020; 13:17. [PMID: 33292371 PMCID: PMC7684758 DOI: 10.1186/s13044-020-00091-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/15/2020] [Indexed: 12/12/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is undoubtedly the thyroid cancer histotype with the poorest prognosis. The conventional treatment includes surgery, radiotherapy, and conventional chemotherapy. Surgery should be as complete as possible, securing the airway and ensuring access for nutritional support; the current standard of care of radiotherapy is the intensity-modulated radiation therapy; chemotherapy includes the use of doxorubicin or taxanes (paclitaxel or docetaxel) generally with platin (cisplatin or carboplatin). However, frequently, these treatments are not sufficient and a systemic treatment with kinase inhibitors is necessary. These include multitarget tyrosine kinase inhibitors (Lenvatinib, Sorafenib, Sunitinib, Vandetanib, Axitinib, Pazopanib, Pyrazolo-pyrimidine compounds), single target tyrosine kinase inhibitors (Dabrafenib plus Trametinib and Vemurafenib against BRAF, Gefitinib against EGFR, PPARγ ligands (e.g. Efatutazone), Everolimus against mTOR, vascular disruptors (e.g. Fosbretabulin), and immunotherapy (e.g. Spartalizumab and Pembrolizumab, which are anti PD-1/PD-L1 molecules). Therapy should be tailored to the patients and to the tumor genetic profile. A BRAF mutation analysis is mandatory, but a wider evaluation of tumor mutational status (e.g. by next-generation sequencing) is desirable. When a BRAFV600E mutation is detected, treatment with Dabrafenib and Trametinib should be preferred: this combination has been approved by the Food and Drug Administration for the treatment of patients with locally advanced or metastatic ATC with BRAFV600E mutation and with no satisfactory locoregional treatment options. Alternatively, Lenvatinib, regardless of mutational status, reported good results and was approved in Japan for treating unresectable tumors. Other single target mutation agents with fair results are Everolimus when a mutation involving the PI3K/mTOR pathway is detected, Imatinib in case of PDGF-receptors overexpression, and Spartalizumab in case of PD-L1 positive tumors. Several trials are currently evaluating the possible beneficial role of a combinatorial therapy in ATC. Since in this tumor several genetic alterations are usually found, the aim is to inhibit or disrupt several pathways: these combination strategies use therapy targeting angiogenesis, survival, proliferation, and may act against both MAPK and PI3K pathways. Investigating new treatment options is eagerly awaited since, to date, even the molecules with the best radiological results have not been able to provide a durable disease control.
Collapse
Affiliation(s)
- Simone De Leo
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Piazzale Brescia, 20, 20149, Milan, Italy.
| | - Matteo Trevisan
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Laura Fugazzola
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Piazzale Brescia, 20, 20149, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
10
|
Sugawara S, Takayanagi M, Honda S, Tatsuta T, Fujii Y, Ozeki Y, Ito J, Sato M, Hosono AM. Catfish egg lectin affects influx and efflux rates of sunitinib in human cervical carcinoma HeLa cells. Glycobiology 2020; 30:802-816. [PMID: 32248228 DOI: 10.1093/glycob/cwaa029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 11/15/2022] Open
Abstract
New treatment protocols are aiming to reduce the dose of the multitargeted tyrosine kinase inhibitor sunitinib, as sunitinib elicits many adverse effects depending on its dosage. Silurus asotus egg lectin (SAL) has been reported to enhance the incorporation of propidium iodide as well as doxorubicin into Burkitt's lymphoma Raji cells through binding to globotriaosylceramide (Gb3) on the cell surface. The objective of this study was to examine whether SAL enhances the cytotoxic effect of sunitinib in Gb3-expressing HeLa cells. Although the treatment with SAL delayed the cell growth and enhanced the propidium iodide uptake, cell death accompanied by membrane collapse was not observed. The viability of sunitinib-treated HeLa cells was significantly reduced when the treatment occurred in combination with SAL compared to their separate usage. Sunitinib uptake significantly increased for 30 min in SAL-treated cells, and this increment was almost completely abolished by the addition of L-rhamnose, a hapten sugar of SAL, but not by D-glucose. After removal of SU from the medium, the intracellular sunitinib level in SAL-treated cells was higher than in untreated cells for 24 h, which was not observed in Gb3-deficient HeLa cells. Furthermore, we observed that SAL promoted the formation of lysosome-like structures, which are LAMP1 positive but not acidic in HeLa cells, which can trap sunitinib. Interestingly, SAL-induced vacuolation in HeLa cells was not observed in another Gb3 positive Raji cells. Our findings suggest that SAL/Gb3 interaction promoted sunitinib uptake and suppressed sunitinib excretion and that sunitinib efficiently exerted cytotoxicity against HeLa cells.
Collapse
Affiliation(s)
- Shigeki Sugawara
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | - Madoka Takayanagi
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan.,Chemiluminescent Reagents Department, R&D Section, Kagamida Factory, DENKA SEIKEN Co. Ltd., 1359-1 Kagamida, Kigoshi Gosen-shi, Niigata 959-1695, Japan
| | - Shota Honda
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | - Takeo Tatsuta
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | - Yuki Fujii
- Graduate School of Pharmaceutical Sciences, Nagasaki International University, 2825-7 Huis Ten Bosch, Sasebo, Nagasaki 859-3298, Japan
| | - Yasuhiro Ozeki
- Department of Life and Environmental System Science, Laboratory of Glycobiology and Marine Biochemistry, Graduate School of NanoBio Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | - Jun Ito
- Department of Urology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Japan
| | - Makoto Sato
- Department of Urology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Japan
| | - And Masahiro Hosono
- Division of Cell Recognition Study, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| |
Collapse
|
11
|
Di Desidero T, Orlandi P, Gentile D, Banchi M, Alì G, Kusmic C, Armanetti P, Cayme GJ, Menichetti L, Fontanini G, Francia G, Bocci G. Pharmacological effects of vinorelbine in combination with lenvatinib in anaplastic thyroid cancer. Pharmacol Res 2020; 158:104920. [PMID: 32461187 PMCID: PMC8011355 DOI: 10.1016/j.phrs.2020.104920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 04/29/2020] [Accepted: 05/10/2020] [Indexed: 12/27/2022]
Abstract
Anaplastic thyroid cancer (ATC) is a rare neoplasia with a poor prognosis. Proliferation and apoptosis assays were performed on ATC cell lines (8305C, 8505C) exposed to vinorelbine, lenvatinib, as well as to concomitant combinations. ABCB1, ABCG2 and CSF-1 mRNA expression was evaluated by real time PCR. The relative levels of pospho Akt were investigated as part of a human phospho-kinase array analysis, and CSF-1 and VEGFR-2 protein levels were measured by ELISA. The intracellular concentration of lenvatinib in ATC cells was measured by combined reversed-phase liquid chromatography-tandem mass spectrometry. An ATC subcutaneous xenograft tumor model in nude mice was treated with vinorelbine, lenvatinib, or vinorelbine plus lenvatinib. After treatment with vinorelbine, lenvatinib, a significant antiproliferative effect in ATC cell lines was observed. The concomitant treatment of vinorelbine and lenvatinib revealed synergism for all the fractions of affected cells. A decrease in ABCB1 expression was reported in both ATC cell lines treated with the lenvatinib plus vinorelbine combination, as was an increase in the intracellular concentration of lenvatinib. The combination caused a decrease in Akt, GSK3α/β, PRAS40 and Src phosphorylation, and in both CSF-1 mRNA and protein levels. In the subcutaneous tumor model, the combination reduced the tumor volume during the treatment period. Our results establish the synergistic ATC antitumor activity of a vinorelbine and lenvatinib combination.
Collapse
Key Words
- Anaplastic thyroid cancer
- Lenvatinib
- Lenvatinib - IUPAC name: 4-[3-chloro-4-(cyclopropylcarbamoylamino)phenoxy]-7-methoxyquinoline-6-carboxamide - PubChem CID: 9823820
- Sorafenib
- Synergism
- Vinorelbine
- Vinorelbine - IUPAC namemethyl (1R, 9R, 10S, 11R, 12R, 19R)-11-acetyloxy-12-ethyl-4-[(12S, 14R,)-16-ethyl-12-methoxycarbonyl-1, 10-diazatetracyclo[12.3.1.0, (3), (11).0, (4), (9),]octadeca-3(11), 4, 6, 8, 15-pentaen-12-yl]-10-hydroxy-5-methoxy-8-methyl-8, 16-diazapentacyclo[10.6.1.0, (1), (9).0, (2), (7).0, (16), (19),]nonadeca-2, 4, 6, 13-tetraene-10-carboxylate- PubChem CID 5311497
Collapse
Affiliation(s)
- Teresa Di Desidero
- Dipartimento Di Medicina Clinica e Sperimentale, Università Di Pisa, Pisa, Italy
| | - Paola Orlandi
- Dipartimento Di Medicina Clinica e Sperimentale, Università Di Pisa, Pisa, Italy
| | - Daniela Gentile
- Dipartimento Di Medicina Clinica e Sperimentale, Università Di Pisa, Pisa, Italy
| | - Marta Banchi
- Dipartimento Di Medicina Clinica e Sperimentale, Università Di Pisa, Pisa, Italy
| | - Greta Alì
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell'Area Critica, Università di Pisa, Pisa, Italy
| | - Claudia Kusmic
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Paolo Armanetti
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Ginelle J Cayme
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| | - Luca Menichetti
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Gabriella Fontanini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell'Area Critica, Università di Pisa, Pisa, Italy
| | - Giulio Francia
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| | - Guido Bocci
- Dipartimento Di Medicina Clinica e Sperimentale, Università Di Pisa, Pisa, Italy
| |
Collapse
|
12
|
Banchi M, Orlandi P, Gentile D, Alì G, Fini E, Fontanini G, Francia G, Bocci G. Synergistic activity of linifanib and irinotecan increases the survival of mice bearing orthotopically implanted human anaplastic thyroid cancer. Am J Cancer Res 2020; 10:2120-2127. [PMID: 32775005 PMCID: PMC7407355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 06/27/2020] [Indexed: 06/11/2023] Open
Abstract
Anaplastic thyroid cancer (ATC) is the most aggressive form of thyroid cancer, and novel combined therapies are urgently needed to prolong patient survival. No data are currently available on the preclinical activity of the combination of linifanib, a CSF-1R inhibitor, and irinotecan in ATC. The aim of the study was to evaluate the in vitro and in vivo activity of linifanib plus irinotecan. Proliferation and apoptosis assays were performed on 8305C and 8505C human ATC cell lines exposed to SN-38, the active metabolite of irinotecan, linifanib alone, and their concomitant combination. Synergism was evaluated by the combination index method. Quantification of pospho-CSF-1R levels was performed by ELISA. In vivo ATC orthotopic xenografts were treated with the single drugs, or their combination, to evaluate their impact on survival. Histology and immunohistochemistry were performed on ATC tissue samples. Both SN-38 and linifanib inhibited in vitro the proliferation of 8305C and 8505C cells in a concentration-dependent manner, whereas their concomitant treatment revealed a strong synergism in the ATC cells. A significant pro-apoptotic activity was found in both ATC cell lines treated with linifanib alone and in combination with SN-38. Moreover, linifanib significantly decreased the levels of phospho-CSF-1R after 24 h and 72 h in both 8505C and 8305C cells, and this was also observed with the concomitant administration of SN-38. In vivo, the combination of linifanib and irinotecan produced a greater survival result than either monotherapy, and resulted in a significant higher median survival. In some of the mice the combination produced a complete response with a macroscopic disappearance of the disease, as confirmed by histology. In conclusion, the synergistic ATC antitumor activity of linifanib/irinotecan combination significantly increased the survival of ATC affected mice and induced some complete responses, suggesting a potential role of this schedule in ATC patient's treatment.
Collapse
Affiliation(s)
- Marta Banchi
- Dipartimento di Medicina Clinica e Sperimentale, Università di PisaPisa, Italy
| | - Paola Orlandi
- Dipartimento di Medicina Clinica e Sperimentale, Università di PisaPisa, Italy
| | - Daniela Gentile
- Dipartimento di Medicina Clinica e Sperimentale, Università di PisaPisa, Italy
| | - Greta Alì
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell’Area Critica, Università di PisaPisa, Italy
| | - Elisabetta Fini
- Dipartimento di Medicina Clinica e Sperimentale, Università di PisaPisa, Italy
| | - Gabriella Fontanini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell’Area Critica, Università di PisaPisa, Italy
| | - Giulio Francia
- Border Biomedical Research Center, University of Texas at El PasoEl Paso, TX, USA
| | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, Università di PisaPisa, Italy
| |
Collapse
|
13
|
Fallahi P, Ferrari SM, Elia G, Ragusa F, Patrizio A, Paparo SR, Marone G, Galdiero MR, Guglielmi G, Foddis R, Cristaudo A, Antonelli A. Primary cell cultures for the personalized therapy in aggressive thyroid cancer of follicular origin. Semin Cancer Biol 2020; 79:203-216. [PMID: 32569821 DOI: 10.1016/j.semcancer.2020.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/27/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022]
Abstract
Thyroid cancer (TC) is the most prevalent endocrine malignancy. More than 90 % of TC is represented by differentiated TC (DTC) arising from the follicular thyroid cells. DTC includes papillary TC (PTC), follicular TC (FTC), and Hürthle cell TC. Anaplastic TC (ATC) accounts for 1% of TC, and it represents 15-40 % of TC death. Current treatment strategies are not completely effective against aggressive DTC or ATC, and mortality is one of the most important challenges. Recently, progresses have been obtained in the understanding of the molecular/genetic basis of TC progression, and new drugs have been introduced [i.e. tyrosine kinase inhibitors (TKIs)], able to block the oncogenic or signaling kinases, associated with cellular growth. Thyroid cell lines, obtained from tumoral cells and chosen for high proliferation in vitro, have been used as preclinical models. Actually, these cells lose the characteristic features of the primary tumor, because they adapt to in vitro growth conditions. For these reasons, the use of these cell lines has important limitations, and more recently human primary cell cultures have been established as monolayer cultures, and investigated for their biological behavior. Moreover, in the past, primary TC cells could be collected only through surgical biopsies, while recently human primary cell cultures can be established also from samples of fine-needle aspiration citology from aggressive dedifferentiated DTC or ATC. Testing in vitro different TKIs in each patient can help to develop new personalized treatments, without using ineffective drugs. In conclusion, personalized medicine and precise oncology, which consider both patients and their disease features, represent the future of the treatment approach, and further progress is needed in this direction.
Collapse
Affiliation(s)
- Poupak Fallahi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Giusy Elia
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesca Ragusa
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Armando Patrizio
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; Center for Basic and Clinical Immunology Research, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization Center of Excellence, University of Naples Federico II, 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council, 80131 Naples, Italy
| | - Maria Rosaria Galdiero
- Center for Basic and Clinical Immunology Research, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization Center of Excellence, University of Naples Federico II, 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council, 80131 Naples, Italy; Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Giovanni Guglielmi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Rudy Foddis
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alfonso Cristaudo
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessandro Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| |
Collapse
|
14
|
Lin B, Lu B, Hsieh IY, Liang Z, Sun Z, Yi Y, Lv W, Zhao W, Li J. Synergy of GSK-J4 With Doxorubicin in KRAS-Mutant Anaplastic Thyroid Cancer. Front Pharmacol 2020; 11:632. [PMID: 32477122 PMCID: PMC7239034 DOI: 10.3389/fphar.2020.00632] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Background Anaplastic thyroid cancer is the most aggressive thyroid cancer and has a poor prognosis. At present, there is no effective treatment for it. Methods Here, we used different concentrations of GSK-J4 or a combination of GSK-J4 and doxorubicin to treat human Cal-62, 8505C, and 8305C anaplastic thyroid cancer (ATC) cell lines. The in vitro experiments were performed using cell viability assays, cell cycle assays, annexin-V/PI binding assays, Transwell migration assays, and wound-healing assays. Tumor xenograft models were used to observe effects in vivo. Results The half maximal inhibitory concentration (IC50) of GSK-J4 in Cal-62 cells was 1.502 μM, and as the dose of GSK-J4 increased, more ATC cells were blocked in the G2-M and S stage. The combination of GSK-J4 and doxorubicin significantly increased the inhibitory effect on proliferation, especially in KRAS-mutant ATC cells in vivo (inhibition rate 38.0%) and in vitro (suppresses rate Fa value 0.624, CI value 0.673). The invasion and migration abilities of the KRAS-mutant cell line were inhibited at a low concentration (p < 0.05). Conclusions The combination of GSK-J4 with doxorubicin in KRAS-mutant ATC achieved tumor-suppressive effects at a low dose. The synergy of the combination of GSK-J4 and doxorubicin may make it an effective chemotherapy regimen for KRAS-mutant ATC.
Collapse
Affiliation(s)
- Bo Lin
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bing Lu
- Institute of Urology of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - I-Yun Hsieh
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen Liang
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Zicheng Sun
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Yi
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Weiming Lv
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Zhao
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| | - Jie Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Li W, Zhan M, Quan YY, Wang H, Hua SN, Li Y, Zhang J, Lu L, Cui M. Modulating the tumor immune microenvironment with sunitinib malate supports the rationale for combined treatment with immunotherapy. Int Immunopharmacol 2020; 81:106227. [DOI: 10.1016/j.intimp.2020.106227] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/17/2020] [Accepted: 01/17/2020] [Indexed: 10/25/2022]
|
16
|
Laetitia G, Sven S, Fabrice J. Combinatorial Therapies in Thyroid Cancer: An Overview of Preclinical and Clinical Progresses. Cells 2020; 9:E830. [PMID: 32235612 PMCID: PMC7226736 DOI: 10.3390/cells9040830] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Accounting for about 2% of cancers diagnosed worldwide, thyroid cancer has caused about 41,000 deaths in 2018. Despite significant progresses made in recent decades in the treatment of thyroid cancer, many resistances to current monotherapies are observed. In our complete review, we report all treatments that were tested in combination against thyroid cancer. Many preclinical studies investigating the effects of inhibitors of the MAPK and PI3K pathways highlighted the importance of mutations in such signaling pathways and their impacts on the subsequent efficacy of targeted therapies, thus reinforcing the need of more personalized therapeutic strategies. Our review also points out the multiple possibilities of combinatory strategies, particularly using therapies targeting proliferation, survival, angiogenesis, and in combination with conventional treatments such as chemotherapies. In any case, resistances to anticancer therapies always develop through the activation of alternative signaling pathways. Combinatory treatments aim to blockade such mechanisms, which are gradually decrypted, thus offering new perspectives for the future. The preclinical and clinical aspects of our review allow us to have a global opinion of the different therapeutic options currently evaluated in combination and to be aware about new perspectives of treatment of thyroid cancer.
Collapse
Affiliation(s)
- Gheysen Laetitia
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine, Mons University, Avenue du Champ de Mars, 8, B7000 Mons, Belgium; (S.S.); (J.F.)
| | | | | |
Collapse
|
17
|
Gentile D, Orlandi P, Banchi M, Bocci G. Preclinical and clinical combination therapies in the treatment of anaplastic thyroid cancer. Med Oncol 2020; 37:19. [DOI: 10.1007/s12032-020-1345-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 02/12/2020] [Indexed: 12/30/2022]
|
18
|
Di Desidero T, Orlandi P, Gentile D, Bocci G. Effects of Pazopanib Monotherapy vs. Pazopanib and Topotecan Combination on Anaplastic Thyroid Cancer Cells. Front Oncol 2019; 9:1202. [PMID: 31799182 PMCID: PMC6863333 DOI: 10.3389/fonc.2019.01202] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 10/22/2019] [Indexed: 12/22/2022] Open
Abstract
The purpose of this study was to examine pazopanib/topotecan combination activity vs. pazopanib monotherapy on anaplastic thyroid cancer (ATC) cells. Proliferation analyses were performed on ATC cell lines administered for 72 h with pazopanib and topotecan alone and to their simultaneous combination. Pazopanib and topotecan produced a strong synergism on ATC cells, calculated by the combination index, increasing the intracellular concentrations of topotecan lactone measured by high-performance liquid chromatography. Furthermore, a significantly decrease of the gene expression of ATP-binding cassette transporter G2 (ABCG-2), vascular endothelial growth factor (VEGF), hypoxia-inducible factor-1α (HIF-1α), and colony stimulating factor-1 (CSF-1) was presented in combination-treated ATC cells by real time PCR tests. In summary, the simultaneous association of pazopanib and topotecan established a highly synergistic ATC antiproliferative effect, suggesting a new possibility to translate this schedule into clinical trials.
Collapse
Affiliation(s)
- Teresa Di Desidero
- Dipartimento di Medicina Clinica e Sperimentale, University of Pisa, Pisa, Italy
| | - Paola Orlandi
- Dipartimento di Medicina Clinica e Sperimentale, University of Pisa, Pisa, Italy
| | - Daniela Gentile
- Dipartimento di Medicina Clinica e Sperimentale, University of Pisa, Pisa, Italy
| | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, University of Pisa, Pisa, Italy
| |
Collapse
|
19
|
Jiang L, Wang L, Zhang Z, Wang Z, Wang X, Wang S, Luan X, Xia Y, Liu Y. The pharmacokinetic interaction between irinotecan and sunitinib. Cancer Chemother Pharmacol 2019; 85:443-448. [DOI: 10.1007/s00280-019-03985-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022]
|
20
|
Abstract
Twenty-five years ago, the cytotoxic drug irinotecan (IRT) was first approved in Japan for the treatment of cancer. For more than two decades, the IRT prodrug has largely contributed to the treatment of solid tumors worldwide. Nowadays, this camptothecin derivative targeting topoisomerase 1 remains largely used in combination regimen, like FOLFIRI and FOLFIRINOX, to treat metastatic or advanced solid tumors, such as colon, gastric and pancreatic cancers and others. This review highlights recent discoveries in the field of IRT and its derivatives, including analogues of the active metabolite SN38 (such as FL118), the recently approved liposomal form Nal-IRI and SN38-based immuno-conjugates currently in development (such as sacituzumab govitecan). New information about the IRT mechanism of action are presented, including the discovery of a new protein target, the single-stranded DNA-binding protein FUBP1. Significant progress has been made also to better understand and manage the main limiting toxicities of IRT, chiefly neutropenia and diarrhea. The role of drug-induced inflammation and dysbiosis is underlined and strategies to limit the intestinal toxicity of IRT are discussed (use of β-glucuronidase inhibitors, plant extracts, probiotics). The detailed knowledge of the metabolism of IRT has enabled the identification of potential biomarkers to guide patient selection and to limit drug-induced toxicities, but no robust IRT-specific therapeutic biomarker has been approved yet. IRT is a versatile chemotherapeutic agent which combines well with a variety of anticancer drugs. It offers a large range of drug combinations with cytotoxic agents, targeted products and immuno-active biotherapeutics, to treat a variety of advanced solid carcinoma, sarcoma and cancers with progressive central nervous system diseases. A quarter of century after its first launch, IRT remains an essential anticancer drug, largely prescribed, useful to many patients and scientifically inspiring.
Collapse
|
21
|
Ferrari SM, Antonelli A, Guidi P, Bernardeschi M, Scarcelli V, Fallahi P, Frenzilli G. Genotoxicity Evaluation of the Soybean Isoflavone Genistein in Human Papillary Thyroid Cancer Cells. Study of Its Potential Use in Thyroid Cancer Therapy. Nutr Cancer 2019; 71:1335-1344. [PMID: 31017483 DOI: 10.1080/01635581.2019.1604004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 12/19/2022]
Abstract
Genistein is one of the several known isoflavonic phytoestrogens found in a number of plants, with soybeans and soy products being the primary food source. The aim of the study is to evaluate if genistein is able to exert antineoplastic action in primary human papillary thyroid cancer (PTC) cells. Thyroid tissues were treated with genistein (1-10-50-100 µM). Cell viability, proliferation, DNA primary damage and chromosomal damage were evaluated. An antiproliferative effect was induced by the highest doses of genistein, and such an effect was synergistically enhanced by the cotreatment with the antineoplastic drug sorafenib. Comet assay did not show any genotoxic effect in terms of primary DNA damage at all the times (4 and 24 h) and tested doses. A reduction of hydrogen peroxide-induced DNA primary damage in primary thyrocytes from PTC cells pretreated with genistein was observed. Data suggest that genistein exerts antineoplastic action, does not induce genotoxic effects while reduces oxidative-induced DNA damage in primary thyrocytes from PTC cells, supporting its possible use in therapeutic intervention.
Collapse
Affiliation(s)
- S M Ferrari
- Department of Clinical and Experimental Medicine - Section of Internal Medicine, University of Pisa , Pisa , Italy
| | - A Antonelli
- Department of Clinical and Experimental Medicine - Section of Internal Medicine, University of Pisa , Pisa , Italy
| | - P Guidi
- Department of Clinical and Experimental Medicine - Section of Applied Biology and Genetics, University of Pisa , Pisa , Italy
| | - M Bernardeschi
- Department of Clinical and Experimental Medicine - Section of Applied Biology and Genetics, University of Pisa , Pisa , Italy
| | - V Scarcelli
- Department of Clinical and Experimental Medicine - Section of Applied Biology and Genetics, University of Pisa , Pisa , Italy
| | - P Fallahi
- Department of Clinical and Experimental Medicine - Section of Internal Medicine, University of Pisa , Pisa , Italy
| | - G Frenzilli
- Department of Clinical and Experimental Medicine - Section of Applied Biology and Genetics, University of Pisa , Pisa , Italy
| |
Collapse
|
22
|
Trabectedin and irinotecan combination regresses a cisplatinum-resistant osteosarcoma in a patient-derived orthotopic xenograft nude-mouse model. Biochem Biophys Res Commun 2019; 513:326-331. [PMID: 30955860 DOI: 10.1016/j.bbrc.2019.03.191] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022]
Abstract
Recurrent osteosarcoma is a chemotherapy-resistant disease. Individualized precision therapy is needed for this disease. Toward this goal, we have developed the patient-derived othotopic xenograft (PDOX) mouse model of all major cancer types including osteosarcoma. Synergistic efficacy of trabectedin (TRAB) and irinotecan (IRT) has been reported in Ewing's sarcoma, soft-tissue sarcoma, and ovarian cancer. However, the efficacy of this combination on osteosarcoma is not known. The goal of present study was to determine the efficacy of the TRAB and IRT combination on cisplatinum (CDDP)-resistant osteosarcoma PDOX. The osteosarcoma PDOX models were randomized into five treatment groups of six mice: Untreated control; CDDP alone; TRAB alone; IRT alone; and TRAB and the IRT combination. Tumor size and body weight were measured during the 14 days of treatment. Tumor growth was regressed only by the TRAB-IRT combination. Tumors treated with the TRAB-IRT combination had the most tumor necrosis with degenerative change. The present study demonstrates the power of the PDOX model to identify a novel effective treatment strategy of the TRAB and IRT combination for chemotherapy-resistant osteosarcoma.
Collapse
|
23
|
Di Desidero T, Orlandi P, Fioravanti A, Alì G, Cremolini C, Loupakis F, Gentile D, Banchi M, Cucchiara F, Antoniotti C, Masi G, Fontanini G, Falcone A, Bocci G. Chemotherapeutic and antiangiogenic drugs beyond tumor progression in colon cancer: Evaluation of the effects of switched schedules and related pharmacodynamics. Biochem Pharmacol 2019; 164:94-105. [PMID: 30953637 DOI: 10.1016/j.bcp.2019.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/01/2019] [Indexed: 11/17/2022]
Abstract
The aim of the study was to evaluate the effects and the related pharmacological mechanisms of switched schedules of antiangiogenic and chemotherapeutic drugs beyond progression after a first-line treatment in a colorectal cancer preclinical model. In vivo studies were performed in nude mice subcutaneously transplanted with colon cancer cells. The treatments included drug combinations with a switch between chemotherapeutic (i.e., irinotecan and 5-fluorouracil) and/or antiangiogenic drugs (i.e., anti-VEGF antibodies and sunitinib) at the time of tumor progression. Proliferation assays were also achieved in vitro on different colon cancer cell lines exposed to SN-38 and sunitinib alone or in combination. ABCG2 gene expression was performed with real-time PCR and SN-38 intracellular concentrations were measured. The switch in the combined treatments, at the time of tumor progression, of the chemotherapeutic (from irinotecan to 5-fluoruracil), or the antiangiogenic drug (from anti-VEGF antibodies to sunitinib) or of both drugs induced a new response. Immunohistochemistry of stromal PDGF-C, PlGF, SD1-α, Tie-2, and VEGFR-2 showed statistical differences between tumors at the time of relapse and after the switched therapy. Moreover, the combination of SN-38 and sunitinib caused synergism on colon cancer cells, with significant inhibition of the ABCG2 gene expression and an increase of SN-38 intracellular concentrations. Our observations may be of clinical relevance, suggesting the switch of single chemotherapeutic or antiangiogenic drugs beyond progression of the disease to obtain a new tumor response due to a modulation of angiogenic factors and a direct effect on tumor cells with a possible variation of intracellular drug concentrations.
Collapse
Affiliation(s)
- Teresa Di Desidero
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Paola Orlandi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Anna Fioravanti
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Greta Alì
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell'Area Critica, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Chiara Cremolini
- U.O. Oncologia Medica 2 Universitaria, Azienda Ospedaliera-Universitaria Pisana, Pisa, Italy; Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Fotios Loupakis
- U.O. Oncologia Medica 2 Universitaria, Azienda Ospedaliera-Universitaria Pisana, Pisa, Italy; Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Daniela Gentile
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Marta Banchi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Federico Cucchiara
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Carlotta Antoniotti
- U.O. Oncologia Medica 2 Universitaria, Azienda Ospedaliera-Universitaria Pisana, Pisa, Italy; Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Gianluca Masi
- U.O. Oncologia Medica 2 Universitaria, Azienda Ospedaliera-Universitaria Pisana, Pisa, Italy; Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Gabriella Fontanini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell'Area Critica, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Alfredo Falcone
- U.O. Oncologia Medica 2 Universitaria, Azienda Ospedaliera-Universitaria Pisana, Pisa, Italy; Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy
| | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Via Savi 10, I-56126 Pisa, Italy.
| |
Collapse
|
24
|
De Martino D, Yilmaz E, Orlacchio A, Ranieri M, Zhao K, Di Cristofano A. PI3K blockage synergizes with PLK1 inhibition preventing endoreduplication and enhancing apoptosis in anaplastic thyroid cancer. Cancer Lett 2018; 439:56-65. [PMID: 30243708 PMCID: PMC6195833 DOI: 10.1016/j.canlet.2018.09.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 09/11/2018] [Accepted: 09/14/2018] [Indexed: 12/18/2022]
Abstract
Anaplastic thyroid cancer (ATC) is among the most lethal malignancies. The mitotic kinase PLK1 is overexpressed in the majority of ATCs and PLK1 inhibitors have shown preclinical efficacy. However, they also cause mitotic slippage and endoreduplication, leading to the generation of tetraploid, genetically unstable cell populations. We hypothesized that PI3K activity may facilitate mitotic slippage upon PLK1 inhibition, and thus tested the effect of combining PLK1 and PI3K inhibitors in ATC models, in vitro and in vivo. Treatment with BI6727 and BKM120 resulted in a significant synergistic effect in ATC cells, independent of the levels of AKT activity. Combination of the two drugs enhanced growth suppression at doses for which the single drugs showed no effect, and led to a massive reduction of the tetraploid cells population. Furthermore, combined treatment in PI3Khigh cell lines showed a significant induction of apoptosis. Finally, combined inhibition of PI3K and PLK1 was extremely effective in vivo, in an immunocompetent allograft model of ATC. Our results demonstrate a clear therapeutic potential of combining PLK1 and PI3K inhibitors in anaplastic thyroid tumors.
Collapse
Affiliation(s)
- Daniela De Martino
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Emrullah Yilmaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Arturo Orlacchio
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Michela Ranieri
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ke Zhao
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Antonio Di Cristofano
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
25
|
Zong S, Li J, Yang L, Huang Q, Ye Z, Hou G, Ye M. Synergistic antitumor effect of polysaccharide from Lachnum sp. in combination with cyclophosphamide in hepatocellular carcinoma. Carbohydr Polym 2018; 196:33-46. [PMID: 29891303 DOI: 10.1016/j.carbpol.2018.05.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/28/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023]
Abstract
Combination therapy with chemotherapeutics is attracting increasing attention as an important treatment option for hepatocellular carcinoma (HCC) due to its complex pathological characteristics. In this study, as a new therapy strategy, combination treatment of LEP-2a (a non-toxic polysaccharide from Lachnum sp.) with cyclophosphamide (CTX) was investigated. Results showed that combination treatment with LEP-2a and CTX processed a significantly synergistic anti-tumor effect in H22 tumor-bearing mice through Fas/FasL mediated caspase-dependent death pathway and mitochondria apoptosis pathway. Moreover, our study indicated that LEP-2a played a crucial role in enhancement of immune response, inhibition of tumor angiogenesis and down-regulation of survival associated proteins. Notably, side effects induced by CTX were relieved after LEP-2a treatment. These results support the conception that LEP-2a has the potential as an ideal adjuvant agent for a more effective combination therapy with CTX against HCC.
Collapse
Affiliation(s)
- Shuai Zong
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Jinglei Li
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Liu Yang
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Qianli Huang
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Ziyang Ye
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Guohua Hou
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Ming Ye
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China.
| |
Collapse
|